You are on page 1of 16

JID: JOINTM

ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

Journal of Intensive Medicine xxx (xxxx) xxx

Contents lists available at ScienceDirect

Journal of Intensive Medicine


journal homepage: www.elsevier.com/locate/jointm

Review

Antimicrobial stewardship and targeted therapies in the changing


landscape of maternal sepsis
Nishel M Shah 1,∗, Esmita Charani 2,3, Damien Ming 4, Fook-Choe Cheah 5, Mark R Johnson 1
1
Department of Metabolism, Digestion and Reproduction, Imperial College London, Chelsea and Westminster Hospital, London, UK
2
Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, UK
3
Division of Infectious Diseases and HIV Medicine, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
4
Department of Infectious Diseases, Imperial College London, Chelsea and Westminster Hospital, London, UK
5
Department of Paediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia

a r t i c l e i n f o a b s t r a c t

Managing Editor: Jingling Bao Pregnant and postnatal women are a high-risk population particularly prone to rapid progression to sepsis with
significant morbidity and mortality worldwide. Moreover, severe maternal infections can have a serious detrimen-
Keywords:
Sepsis tal impact on neonates with almost 1 million neonatal deaths annually attributed to maternal infection or sepsis.
Pregnancy In this review we discuss the susceptibility of pregnant women and their specific physiological and immunolog-
Immunology ical adaptations that contribute to their vulnerability to sepsis, the implications for the neonate, as well as the
Cardiovascular issues with antimicrobial stewardship and the challenges this poses when attempting to reach a balance between
Personalised medicine clinical care and urgent treatment. Finally, we review advancements in the development of pregnancy-specific
Antibiotic stewardship diagnostic and therapeutic approaches and how these can be used to optimize the care of pregnant women and
neonates.

Introduction ments in care. Age standardized infection rates have fallen from
800 per 100,000 pregnancies in 1990 to 550 per 100,000 preg-
Sepsis has a significant contribution to both global morbidity nancies in 2019.[ 5 ] Despite this, in 2019 there were over 20
and mortality with approximately 30 million patients affected million cases of maternal sepsis and other maternal infections
worldwide each year and a resulting 6 million deaths.[ 1 , 2 ] Poor worldwide. The World Health Organization (WHO) reported
outcomes from sepsis are a particular problem in certain vulner- global prevalence of maternal sepsis is 4.4%[ 5 , 6 ] . In the UK,
able populations which includes pregnant and postnatal women. significant risk factors include ethnicity and deprivation. Black
One in 10 deaths in pregnant and postnatal women worldwide and other ethnic minority groups are at an almost 2-fold in-
is attributed to sepsis[ 2 , 3 ] and one million neonatal deaths are creased risk.[ 7 , 8 ] Furthermore, when compared to age-matched
secondary to infection in pregnant women, including sepsis.[ 2 , 4 ] non-pregnant controls, sepsis-related case fatality is higher dur-
Physiological adaptations during pregnancy are thought to con- ing pregnancy, childbirth, and postpartum.[ 9 ] The reasons for
tribute to their vulnerability. In particular, changes in their car- the higher rates of sepsis-related maternal morbidity and mor-
diorespiratory and immune systems, alongside physical, hor- tality are not fully understood.
monal, and immunological changes during labor and postpar-
tum have been shown to have a significant impact. Evidence for poor Outcomes from sepsis in pregnancy

Are Pregnant Women at Greater Risk of Sepsis? Data from current and previous coronavirus pandemics as
well as influenza and Ebola demonstrate the increased risk of
Morbidity and mortality from sepsis in pregnancy worldwide severe infection during pregnancy and poorer outcomes. [ 10–14 ]
have gradually been falling over the decades due to improve- For example, in pregnancy the risk of viral pneumonia and


Corresponding author: Nishel M Shah, Department of Metabolism, Digestion and Reproduction, Imperial College London, Chelsea and Westminster Hospital,
London SW10 9NH, UK.
E-mail address: nishel.shah@imperial.ac.uk (N.M. Shah).

https://doi.org/10.1016/j.jointm.2023.07.006
Received 6 April 2023; Received in revised form 4 July 2023; Accepted 30 July 2023. Managing Editor: Jingling Bao
Available online xxx
Copyright © 2023 The Author(s). Published by Elsevier B.V. on behalf of Chinese Medical Association. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/)
Please cite this article as: N.M. Shah, E. Charani, D. Ming et al., Antimicrobial stewardship and targeted therapies in the changing landscape of
maternal sepsis, Journal of Intensive Medicine, https://doi.org/10.1016/j.jointm.2023.07.006
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

subsequent respiratory complications is far greater when com- are important tools to improve poor outcomes. In humans, cer-
pared to the general population.[ 15 ] During the coronavirus 2 tain pathogens predominate. In maternal sepsis, Escherichia coli
(SARS-CoV-2) pandemic, pregnant patients with severe acute (E. coli) and group B Streptococcus (GBS) are the most com-
respiratory syndrome were more likely to require intensive care mon bacterial pathogens, although other causative organisms
and mechanical ventilation, and their mortality rate was higher include other Gram-negative organisms such as Klebsiellas, My-
when compared to non-pregnant women.[ 12 ] Other pathology coplasma, and other coliforms, but also Gram-positive organisms
such as renal failure and disseminated intravascular coagulopa- such as Clostridium sordellii.[ 27 ] However, the most severe out-
thy (DIC) have also been shown to be more frequent in preg- comes are associated with E. coli and group A Streptococcus
nancy. These vascular changes appear to extend to the placenta (GAS).[ 19 ] Progression to septic shock is particularly a problem
where findings consistent with abnormal blood flow and ex- with GAS infection.[ 28 ] Furthermore, in pregnancy, the risk of
tensive thrombotic vasculopathy have been observed in women group A and group B streptococcal bacteremia when compared
delivered during acute infection.[ 14 ] During antenatal bacterial to non-pregnant women is 20–100-fold higher.[ 29 , 30 ]
sepsis, obstetric complications associated with abnormal utero-
placental blood flow such as fetal growth restriction, placen- Why are Pregnant Women at Greater Risk of Sepsis?
tal abruption, and preterm birth are more common.[ 16 ] This
has also been shown in lipopolysaccharide (LPS) treated rats, Physiological changes in pregnancy such as immunological,
simulating sepsis during pregnancy, where coagulopathy, struc- cardiovascular and respiratory adaptations (Figure 1) may con-
tural abnormalities in the uteroplacental vasculature, and de- tribute to an increased risk of developing severe complications
creased placental blood flow lead to fetal hypoxia and preg- during sepsis.
nancy loss.[ 17 ]
Immunological
Period of greatest risk and associative factors in pregnancy
Peripheral blood adaptations
The stage of pregnancy and postpartum is also important. The earliest hypothesis proposed in the 1950′s described the
Almost 34% of all maternal infections occur in the antenatal pe- immune adaptations of pregnancy as a series of changes de-
riod. Moreover, of those infections that are complicated or se- signed to prevent rejection of the semi-allogenic fetus result-
vere, 17% and 28% respectively occur in the antenatal period. ing in maternal immune tolerance.[ 31–33 ] Subsequently, the clas-
In comparison, 60% of all maternal infections occur during the sic model of immune tolerance included a shift in T-helper
intrapartum and postnatal periods, and they account for 60% of subsets (Th1 to Th2) that favored an anti-inflammatory pro-
both complicated and severe infections.[ 18 ] Prospective studies file but also increased susceptibility to infections in pregnancy.
conducted in the UK suggest that labor and the puerperium carry Our current understanding is that the immune environment
a much higher risk, approaching a 2–3-fold increase in risk when transitions with advancing gestation from a pro- to anti- and
compared to the antenatal period.[ 19 ] It is possible that peripar- back to a pro-inflammatory state.[ 34–36 ] The innate and adaptive
tum and postnatal invasive interventions such as Caesarean sec- arms of the immune response are thus differentially activated to
tion and instrumental delivery, as well as intrauterine or vaginal modulate these changes as well as compensate for each other.
tamponade for massive obstetric haemorrhage, increase the risk Immune modulation is thought to be influenced and main-
of developing infection.[ 7 , 20 , 21 ] These interventions have been tained by endogenous factors that include feto-placental de-
the target of efforts to reduceis infection rates by changes in ob- rived antigens and pregnancy hormones such as oestrogen and
stetric care and optimized use of antibiotic prophylaxis.[ 22 , 23 ] progesterone.[ 37–39 ] Fetal antigens elicit tolerogenic humoral
The promotion of aseptic techniques and antibiotics are of par- and cellular responses in pregnant women through the gener-
ticular need in Low and Low Middle Income Countries (LMIC), ation of antigen specific CD4 and CD8 T cells, MHC-specific B
where rates of maternal infections and severe maternal infec- cells (in murine models), and fetal-specific CD4 T-regulatory
tions are 40% and 20% greater respectively than upper-middle cells (Tregs). [ 40–42 ] The placenta is the likely source of fetal
income countries, and fatality following puerperal infection is antigen exposure that is shed through microvesicles and exo-
as high as 50%.[ 6 , 18 ] somes, cellular debris and due to microchimerism.[ 43–47 ] Pro-
As discussed above, severe respiratory complications are gesterone is a key hormonal modulator of immune responses
more common in pregnancy during severe infection, and so are in pregnancy that acts to suppress inflammatory and cytotoxic
other downstream effects such as DIC, renal failure, and hy- functions.[ 36 , 48–50 ]
potension, which contribute to the rapid progression to sep- Altogether, these changes described above result in a dy-
tic shock. In a mouse model of LPS induced sepsis response, namic immune system in pregnancy that varies throughout ges-
pregnancy was found to be associated with a profound hy- tation. In early pregnancy, we see T cells resembling a Th1 phe-
potensive response and increased mortality when compared notype and Tregs that favor an effector subset.[ 51 , 52 ] As preg-
to non-pregnant mice.[ 24 ] In a second series of experiments, nancy progresses into the second trimester the immune envi-
using a polymicrobial sepsis model caused by caecal ligation ronment becomes more immune tolerant. Thus, effector mem-
and puncture the same research group found that pregnancy ory T cells predominate with upregulation of inhibitory mark-
was again associated with a marked hypotensive response and ers, increased progesterone sensitivity of natural killer (NK)
much greater mortality.[ 25 ] Interestingly, the use of broad- cells, altered dendritic cell (DC) subsets and indoleamine 2,3-
spectrum antibiotics and a vascular smooth muscle specific dioxygenase activity, and a shift in Treg phenotype from ef-
inhibitor of nitric oxide synthesis was associated with better fector to naive.[ 35 , 36 , 52 ] Finally, in the third trimester there
outcomes.[ 26 ] This suggests that identification and treatment is a move to favor immune activation with pro-inflammatory

2
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

Figure 1. Physiological adaptations in pregnancy. The respiratory, cardiovascular, and renal changes that occur in pregnancy. Immune changes are subdivided into
systemic and local changes that enable fetal tolerance. These adaptations contribute to the increased risk of maternal sepsis.
ERV: Expiratory reserve volume; FRC: Functional residual capacity; GFR: Glomerular filtration rate; ILC: Innate lymphoid cells; NK: Natural killer.

antigen-specific cellular responses that are further enhanced which produces Interleukin (IL)-22 and IL-17. In the decidua
during labor.[ 36 , 38 ] during pregnancy, ILC2 is thought to be the most abundant sub-
set present.[ 58 ] However, ILC1 and ILC3 may have key roles dur-
Immune adaptations at the maternal-fetal interface ing certain pregnancy pathology, including preterm labor (PTL),
Modulation of immune responses is also evident locally at as well as during physiological labor. Furthermore, during preg-
the maternal-fetal interface and is seen in both the innate and nancy, the composition of these cells in the decidua basalis and
adaptive arms that include NK cells, innate lymphoid cells parietalis is different and may represent a varied immune re-
(ILCs), and T cells. Importantly, this interface tolerates the semi- sponse. For example, Mendes et al.[ 59 ] demonstrated an increase
allogeneic fetus but can retain an ability to protect against in- in ILC2 and ILC3 populations in the decidua basalis in PTL, and
vading pathogens. In fact, 30–40% of decidual cells are leuko- similarly, Xu et al.[ 58 ] saw increases in both basalis and pari-
cytes and this forms a specialized decidual immune system in etalis.
pregnancy.[ 53 ] Uterine NK cells (uNK) from a large portion of Tissue resident macrophages are situated close to decidual
innate leukocytes in the endometrium and decidua and they natural killer (dNK) cells. They function to clear debris, present
function to facilitate placentation during pregnancy. Crucially, feto-placental antigens and secrete cytokines and chemokines to
the uNK phenotype changes during early pregnancy from proan- modulate dNK and T cell responses.[ 60 , 61 ] Both CD8 and CD4 T
giogenic to cytokine producing, thus allowing for extravillous cells can directly (via HLA-C) or indirectly (via antigen process-
trophoblast invasion.[ 54 , 55 ] Their cytotoxicity is largely limited ing cells or APCs) recognize feto-placental antigens. However,
due to their ability to interact with human leucocyte antigens antigen specificity provides a means to regulate T cell immune
(HLA) class I molecules expressed by trophoblast (HLA-C, HLA- responses to prevent fetal rejection. For example, CD8 T cells ex-
E, HLA-G). With labour onset, the decidual NK phenotype shifts pressing inhibitory markers (T cell immunoglobulin and mucin
resulting in a reduction in proportions of cytokine producing domain-containing protein 3 or Tim-3, Programmed cell death 1
CD56hi NK (our unpublished observations). This is consistent or PD-1) that interact with their ligands (Programmed cell death
with single cell RNA-seq data from the placenta, which has ligand 1 or PD-L1) on trophoblast are enriched in the decidua
demonstrated an upregulation of NK cell signatures.[ 56 ] This in an HLA-C dependent manner.[ 62 ] Moreover, the cytotoxic po-
suggests that the composition of the uNK compartment is dy- tential of the effector memory CD8 T cell subset is impaired.[ 63 ]
namic. Strunz et al.[ 57 ] identified a subset of highly prolifera- Further regulation is provided by decidual fetal-specific Tregs
tive uNK cells that regenerate under the influence of sex hor- that clonally expand during pregnancy.[ 42 ] In murine models,
mones and genetic triggers, for example during menstruation these have been shown to have fetal-specific and highly sup-
and pregnancy. Whilst these cells differentiate locally their ori- pressive capabilities.[ 64 ] In addition, class-switched memory B
gin appears to be from the peripheral circulation and they are cells and IL-10-producing B cells have been found co-localized
transiently tissue-resident. in clusters with Tregs in pregnancy further enhancing the regu-
Another group of tissue resident immune cells important for lation of local immune responses.[ 65 ]
pregnancy is innate lymphoid cells (ILCs), which have been
identified in the uterus and decidua. These have immune modu- Immune contribution to sepsis
latory functions and provide protection against pathogens. They Antenatal responses to infections and vaccines are an op-
are subdivided into group 1 which produces mostly Interferon portunity to understand how pregnancy may affect adaptive
(IFN)-𝛾, group 2 which produces Th2 cytokines, and group 3 immunity. Forbes et al.[ 66 ] showed impaired in vitro antiviral

3
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

IFN responses in the second and third trimesters following rhi- aldosterone (RAA) system that helps to maintain BP. Much
novirus infection. In contrast, analysis of ex vivo responses fol- of this is due to the fact that progesterone, which also rises
lowing influenza vaccine and SARS-CoV-2 infection fare differ- in pregnancy, has potent mineralocorticoid effects, and acts
ently, with maintenance of both humoral and antigen-specific as an aldosterone antagonist to inhibit aldosterone binding.
responses.[ 67–69 ] This suggests that cellular responses are un- This leads to compensatory RAA activation and a rise in
affected by pregnancy immune modulation. However, pheno- serum aldosterone.[ 81 , 82 ] Furthermore, relaxin stimulates vaso-
typic data from the same research studies suggest a reduction pressin secretion causing further water retention. The increase
in the pro-inflammatory cellular compartments and low-level in plasma volume and cardiac output alongside renal vasodilata-
immune regulation.[ 67 , 68 ] With peripartum risks much greater tion, lead to an increase in renal plasma flow and glomerular
than during the antenatal period, labor, which is itself a process filtration rate.
of inflammation, provides an opportunity to understand how Kidney size also increases in pregnancy with physiological
inflammatory responses are regulated. Tissue necrosis because dilatation of the collecting ducts that causes hydronephrosis in
of inflammation can release microparticles to interact with in- most pregnant women. This hydronephrosis increases with ges-
nate immune receptors such as toll-like receptors (TLRs) to drive tation and is greater on the right kidney due to the compressive
inflammatory signals during labor.[ 70 , 71 ] Interestingly, TLR in- effect of the gravid uterus.[ 83 ]
hibitors can repress this output.[ 70 ] As discussed earlier, in preg-
nancy many regulatory immune cells are upregulated, including Cardiovascular contribution to sepsis
Tregs, and these have a suppressive effect on certain immune Increased levels of prostaglandins and nitric oxide (NO),
responses.[ 39 ] Tregs can modify their targets from Th1 to Th2 which are upregulated by oestrogen, encourage smooth muscle
effector T cells alongside a change in their suppressive activ- relaxation and vasodilatation. During sepsis, further upregula-
ity to either suppress or propagate inflammation. Whilst during tion of NO leads to the development of septic shock.[ 84 ] In addi-
sepsis Tregs are thought to have a protective effect by suppress- tion, during sepsis, inflammatory cytokines such as tumor necro-
ing exaggerated inflammatory immune responses, their abun- sis factor (TNF)-𝛼 and IL-1𝛽 lead to myocardial depression.[ 85 , 86 ]
dance in pregnancy may not necessarily be helpful in certain Furthermore, measurable levels of serum Troponin T and I,
circumstances.[ 72–74 ] For example, during labor Treg function and B-type natriuretic peptide (BNP) have been shown to be
is thought to be altered to enable a proinflammatory cytokine increased with sepsis-associated myocardial depression.[ 87–89 ]
response.[ 38 , 39 ] This suggests that immune regulation during the These are useful markers for myocardial function and prognosis
peripartum period may, in fact, result in an augmented inflam- in sepsis. Considering that cardiac output is increased in preg-
matory response to sepsis. In addition to this, leucocyte traffick- nancy, a loss of myocardial function has a significant effect on
ing may also be altered in pregnancy meaning that important the circulatory system. Downstream complications of severe sys-
immune cells are less able to clear pathogens at sites of infec- temic infections include acute kidney injury, which is associated
tion. This is shown in murine work using knockout mice which with significant morbidity and mortality.
demonstrates that the loss of a monocyte chemoattractant re-
Renal contribution to sepsis
ceptor leads to reduced cell trafficking, significantly worse bac-
Physiological hydronephrosis and the mechanical compres-
teremia, and survival.[ 25 ] Furthermore, the migratory potential
sion of the ureters by the gravid uterus cause urinary stasis. This
of important immune cells required during an infection such
alone can increase the risk of bacteriuria and ascending urinary
as neutrophils varies with gestation and during different labor
tract infection in pregnancy by up to 40%.[ 90 ] Furthermore, re-
phenotypes.[ 75 , 76 ] These areas of immune modulation are all
nal tubular epithelial cells can interact with inflammatory me-
possible avenues of future research and treatment options.
diators generated during infections through pattern recognition
receptors (PRRs) such as TLRs.[ 91 ] This interplay can exacerbate
Cardiovascular
local tissue injury. During sepsis, tissue inflammation, microvas-
cular injury, hypoperfusion, and organ dysfunction ensue as a
Circulatory adaptations
cascade of events. Local inflammatory responses form an im-
Cardiac output increases gradually in pregnancy, reach-
portant part of the host’s defense against invading organisms.
ing up to 45% greater than early pregnancy by 24 weeks of
This is mediated by the interaction between pathogen associated
gestation.[ 77 , 78 ] This occurs due to changes in stroke volume
molecular patterns (PAMPs) and danger associated molecular
and heart rate and is associated with both left and right ven-
patterns (DAMPs) released into the vasculature during the initial
tricular hypertrophy. Alongside these changes, peripheral vas-
innate immune response and TLRs found on immune cells.[ 92 ]
cular resistance falls in the second trimester and reaches a nadir
Renal vascular and tubular epithelial cells also express TLRs
of 40%.[ 77 ] Blood pressure (BP) dynamics show similar changes
and their activation leads to the production of reactive oxygen
with a fall in both systolic and diastolic, and therefore mean
species (ROS) and cytokine signaling to attract leucocyte infil-
arterial BP in early pregnancy that falls further in the sec-
trates. The consequent endothelial injury and tubular inflamma-
ond trimester before increasing in the third.[ 79 ] Some of these
tion lead to microthrombi formation, interstitial oedema, and
changes are hormone driven. For example, higher concentra-
further tissue injury.[ 93 ]
tions of progesterone and relaxin are associated with lower sys-
tolic BPs.[ 80 ] Respiratory

Renal adaptations Respiratory adaptations


Plasma volume and red cell mass increase with advancing In pregnancy, there is an increase in oxygen consumption and
gestation. This is due to activation of the renin-angiotensin- progressive distension of the uterus that affects both metabolic

4
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

demands and lung volume. To try to maintain the total lung ca- EOS, commonly by GBS, can have significant neurological se-
pacity, the volume of air remaining in the lungs post-exhalation quelae including seizures, blindness, hearing loss, and speech
measured as functional residual capacity (FRC) and expiratory and language delays.[ 100 ]
reserve volume (ERV) is reduced.[ 94 ] In addition, there is an in- Rates of EOS are higher in some LMICs where the reported
crease in the tidal volume, which results in greater minute ven- incidence of peripartum infection is 4% but symptom reporting
tilation without increasing the respiratory rate. suggests a much more plausible 16%. Early neonatal mortality
due to puerperal sepsis carries a relative risk of 2-fold when
Respiratory contribution to sepsis compared to neonatal deaths in the absence of maternal infec-
The adaptations in pregnancy including the physiological hy- tion. The authors of the study using demographic health survey
perventilation result in a state of respiratory alkalosis. This is data from five LMICs suggest that inadequate reporting of ma-
in part due to progesterone, which increases the sensitivity of ternal sepsis is a significant challenge in the study countries, and
the respiratory center to carbon dioxide.[ 95 , 96 ] To maintain acid- improved recognition and treatment of maternal infections may
base balance, this alkalosis is managed by an increase in renal reduce neonatal complications.[ 102 ]
bicarbonate excretion.[ 94 ] Consequently, the pregnant woman’s
capacity to counter sepsis-related metabolic acidosis may be sig-
Maternal to fetal transmission
nificantly impaired. This is an important consideration for all
forms of sepsis in pregnancy.
During sepsis in pregnancy, vertical transmission to the
During respiratory tract infections, local responses to inflam-
neonate of the causative organism as well as transmission of
mation are important in the context of pregnancy. As discussed,
maternal colonising organisms (such as GBS) is more likely in
pregnant women have been disproportionately affected during
the presence of other infectious morbidities. These include risk
respiratory pandemics. It is possible that these outcomes are
factors such as prolonged rupture of membranes or intrapartum
likely to be made worse by the compressive effect of the gravid
fever that increase the impact and risk of EOS[ 98 ] . Generally, in
uterus on the maternal lungs and the greater chance of basal at-
both these circumstances, the risk of EOS increases 3–4-fold, and
electasis during late pregnancy.[ 94 ] One of the drivers of clinical
50–67% of these involve preterm or LBW neonates.[ 103 ] The pro-
severity in these patients is thought to be a heightened cytokine
gression of ascending lower genital infections will be further ex-
response, which has been observed previously during the H1N1
acerbated by inflammation at the maternal-fetal interface. This
influenza pandemic.[ 97 ] A similar mechanism is thought to be at
causes local immune defenses to be more permeable to invading
play during the COVID-19 pandemic. However, acute changes
organisms, and can also lead to infections overwhelming host
in the lungs may also be affected by pregnancy-specific inflam-
responses.[ 104 , 105 ]
matory leucocyte sequestering in lung tissue during a cytokine
Other potential sources for vertical transmission of infec-
storm, as shown in murine pregnancy sepsis models, eventually
tions are maternal urinary tract infections and respiratory
causing severe tissue damage.[ 24 ]
infections. Urosepsis in pregnancy is a potential source of
What are the Neonatal Consequences of Maternal Sepsis? Gram-negative bacteremia and is associated with poor neonatal
outcomes includingsepticemiaepticaemia, preterm births, and
For the neonate, maternal transmission of bacteria such stillbirths.[ 106 ] Although the mechanism for the relationship is
as GBS or transmission of infections such as chorioamnionitis not clear, the prevalence of neonatal urinary tract infections
during the intrapartum period significantly increases the risk in those born to mothers with a corresponding urinary infec-
of early onset neonatal sepsis (EOS).[ 98 ] In addition, neona- tion is relatively increased.[ 107 , 108 ] Data from LMIC suggest that
tal immune function during PTL with chorioamnionitis is as- neonates born to mothers who experienced urinary tract infec-
sociated with greater inflammatory cytokines and leucocyte tions during pregnancy are 3.55 times more likely to develop
activation.[ 99 ] neonatal sepsis when compared to those born to mothers with-
out a urinary tract infection.[ 109 ] It is plausible that untreated
Early neonatal sepsis ascending urinary infections in pregnancy that lead to sepsis
may result in fetoplacental transmission through hematogenous
Unsurprisingly, rather like maternal sepsis, the most com- spread to infect the neonate.
mon pathogens involved are GBS and E. coli, accounting for ap- Respiratory tract infections are another common source of
proximately 70% of cases of EOS.[ 100 , 101 ] Whilst the overall in- maternal sepsis. Bacterial pneumonia is associated with worse
cidence of EOS is 1–2 per 100 live births, mortality from EOS neonatal outcomes that include preterm birth, low birth weight,
approaches 3% amongst term newborns and 16% in high risk, and pre-eclampsia.[ 15 , 110 ] This is after accounting for comorbidi-
low birth weight (LBW) neonates.[ 98 ] Interestingly, as causative ties such as obesity and diabetes. However, the mechanisms for
agents identified in neonates, GBS appears to affect more term these differences are unknown. While EOS is rare, it remains
neonates and E. coli predominantly preterm infants, with the lat- possible in the presence of maternal bacteremia. Likewise, dur-
ter slowly on the rise. However, other organisms include Strep- ing COVID-19 infection, pregnant women are more likely to de-
tococcus species and Haemophilus influenzae.[ 101 ] velop severe illness with an increased risk of preterm birth and
Significant morbidity from EOS disproportionately af- small for gestational age babies.[ 111–114 ] These neonatal com-
fects preterm neonates, who in the acute phases often plications are more prevalent in moderate to severe COVID-
developing respiratory distress requiring invasive ventilation, 19.[ 115 ] However, early neonatal COVID-19 infection is rare and
and hyper/hypo-glycemia. In the long term, these neonates are neonatal symptoms are generally mild.[ 116 ] The source of ver-
at risk of bronchopulmonary dysplasia (BPD), a poor neurode- tical transmission remains uncertain and appears to be unlike
velopmental outcome, and cerebral palsy. Term neonates with other viruses such as Zika where trophoblast infection serves

5
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

as a route of transmission.[ 117 ] Viral entry is possible through microbiome, and there is evidence that chronic medical prob-
the angiotensin converting enzyme-2 (ACE-2) receptor isolated lems in later life such as childhood asthma may be influenced
on placental tissue and following placental barrier damage sec- by early use of antibiotics.[ 131 ] Currently, however, there are
ondary to local inflammation and tissue injury.[ 118–120 ] Indeed, insufficient data to make clinical recommendations.[ 132 ]
the SARS-CoV-2 virus has been detected in syncytiotrophoblast The development and spread of antimicrobial resistance
and other placental tissues, and the presence of the virus has (AMR) in neonatal populations is also a concern. The WHO
been shown to induce a robust immune response.[ 120–122 ] In 2014 annual report of AMR found that the proportions of E.
placentas with a high viral load, the maternal space, and fe- coli, Klebsiella pneumonia and Staphylococcus aureus resistant to
tal chorionic villi are occupied by extensive inflammatory infil- commonly used antibacterial agents, exceeded 50% in many
trates that comprise macrophages and T cells, and CD56+ NK settings.[ 133 ] In newborns cared for in neonatal units, genes
cells within the decidua.[ 121 ] A similar T cell infiltrate is seen in conferring AMR are detectable in bacteria colonizing the in-
the fetal chorionic plate. Moreover, ACE-2 receptors which are testines of preterm infants. In the same units, Staphylococ-
detectable in fetal kidney, rectum, and ileum, are barely seen in cal species resistant to methicillin and vancomycin are be-
fetal lungs, brain, or heart towards term pregnancy.[ 123 ] In fact, coming more prevalent with a greater mortality associated
surveillance data suggests that postnatal transmission (via res- with infections from these organisms because they are signif-
piratory spread, close contact) accounts for most cases of early icantly more difficult to treat effectively. Furthermore, other
neonatal infections. organisms including extended-spectrum 𝛽-lactamase (ESBL)-
Ultimately the risk of vertical transmission will depend on producing Gram-negative pathogens that are resistant to
the maternal system infected, the quality of maternal immune cephalosporins, Gram-negative bacteria resistant to piperacillin-
response, the receptiveness of the fetal compartment, and the tazobactam, aminoglycosides, and Klebsiella-resistant to car-
evasive nature of the organism in question. bapenems are slowly emerging.[ 134 ] The Neonatal AntiMicro-
bial Resistance (NeoAMR) launched in 2017 with 39 partici-
Management of early neonatal sepsis pating neonatal units across 12 countries, has thus far reported
preliminary data over 12 months, showing AMR rates amongst
National Institute for Health and Care Excellence (NICE) cephalosporins of 26% to 84%, carbapenems of up to 81%, and
guidelines for the management and treatment of neonatal infec- glycopeptide of up to 45%.[ 135 ] These data demonstrate the need
tion (NG195), which includes EOS, comprises empirical treat- for antibiotic stewardship as an increasingly urgent intervention
ment based on maternal risk factors and neonatal clinical re- in neonates.
view at birth.[ 124 ] Thereafter, the duration of treatment is based
on clinical improvement, neonatal blood cultures results, which Is There a Role for Antibiotic Stewardship in Maternal and
have notoriously poor sensitivity, and changes in non-specific Neonatal Clinical Pathways?
inflammatory markers, namely C-reactive protein (CRP).[ 125 , 126 ]
However, in the era of better antibiotic stewardship, the useful- To manage sepsis effectively, clinical pathways have been
ness of empirical treatment based on maternal risk factors in designed to include recognition, resuscitation, and the timely
‘well’ infants is debatable, and tools to determine a baby’s risk administration of appropriate antibiotics.[ 136 ] Guidance is avail-
of developing EOS may be helpful in some cases.[ 125 ] Having able for recommended antibiotic regimes for most obstetric in-
said this, it is important to note that neonatal antibiotic stew- dications as well as EOS (Table 1). The overwhelming message
ardship is mired by challenges, including difficulties in devel- of the surviving sepsis campaigns is that the prompt diagnosis of
oping unit antibiograms, and a lack of robust antibiotic trials sepsis and initiation of broad-spectrum antibiotics is paramount.
in neonatal populations.[ 127 ] There are also challenges with us- Initiatives such as Sepsis Six require that these antibiotics be
ing diagnostic tools in neonatal populations. Research compar- commenced within 3 h, and these recommendations have sig-
ing using the Kaiser Permanente sepsis risk calculator (SRC) to nificantly reduced mortality rates from sepsis.[ 137 , 138 ] One of the
standard NICE guidelines across 8 maternity units in Wales by unintended consequences of the sepsis campaigns has been the
Goel et al.,[ 125 , 128 ] suggests that the use of the tool would have inappropriate use of clinical sepsis pathways in patients who do
resulted in a 55% reduction in neonates receiving antibiotics. not fulfill the criteria for sepsis. Studies have reported that up
However, in a recent meta-analysis of the current published lit- to 50% of patients treated with broad-spectrum antibiotics for
erature surrounding the SRC (including the study by Goel et al.), sepsis, did not have sepsis.[ 139 ]
the authors found that many EOS cases were missed by the tool.
This suggests that whilst the intentions of the SRC are a move in The need for maternal antimicrobial stewardship
the right direction, research investigating the usefulness of such
tools is still in its infancy.[ 129 ] Clearly, a balance is required between effective treatment
and appropriate antibiotic use. Antibiotic stewardship is re-
Complications of antibiotic use in neonates quired to optimize antibiotic usage, including rationalizing and
de-escalating or stopping agents, to maintain their effectiveness.
Whatever the case, there is growing evidence that prolonged This is important because the incidence of AMR and healthcare-
antibiotic use in this population may be detrimental, especially associated infections (HCAIs) is rising.[ 152 ] In Europe, the AMR
in preterm neonates without proven infection, increasing the in 2018 as reported by the European AMR Surveillance Net-
risk of mortality, BPD, necrotizing enterocolitis, retinopathy work (EARS-Net) indicated more than half of the E. coli and
of prematurity, and periventricular white matter damage.[ 130 ] more than a third of the Klebsiella pneumonia isolates were re-
Long-term empirical treatment will also affect the neonatal gut sistant to at least one antibiotic class.[ 153 ] In LMIC these figures

6
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

Table 1
Common obstetric indications for antibiotics and recommended regimes.

Indication Causative organisms Antibiotic recommendations Important notes References

Pregnancy and postnatal for the mother


Antibiotic prophylaxis

GBS prophylaxis GBS Benzylpenicillin Alternative: Cephalosporin Prevention of early-onset neonatal


Penicillin allergy (severe): group B streptococcal disease.
Vancomycin Green-top Guideline No. 36. RCOG
2017 [ 140 ]
Prophylaxis at caesarean Staphylococci Streptococci Single dose first-generation Avoid amoxicillin plus WHO recommendation on
section Gram-negative cephalosporin or penicillin (dose, clavulanate in preterm cases due prophylactic antibiotics for women
30–60 min before surgery) to risk of necrotizing enterocolitis undergoing caesarean section.
Geneva: World Health Organization;
2021 [ 141 ]
Prophylaxis at caesarean Staphylococci Streptococci Single dose first-generation Avoid amoxicillin plus ACOG Practice Bulletin No. 199: Use
section Gram-negative cephalosporin (cefazolin) (dose, clavulanate in preterm cases due of Prophylactic Antibiotics in labour
within 60 min before surgery) to risk of necrotizing and Delivery. Obstet Gynecol. 2018
[ 142 ]
Penicillin allergy: enterocolitis.
Clindamycin + an Azithromycin may be used as an
aminoglycoside adjunct for non-elective
caesareans
Prophylaxis for operative Gram-negative anaerobes Single dose of amoxicillin and As soon as possible after birth WHO recommendation on routine
vaginal birth Streptococci clavulanic acid and no more than 6 h after birth antibiotic prophylaxis for women
undergoing operative vaginal birth.
Geneva: World Health Organization;
2021 [ 143 ]
Assisted vaginal birth. Green-top
Guideline No. 26. RCOG, 2020 [ 144 ]

Antibiotic treatment for suspected sepsis

Intrapartum pyrexia Streptococci Pyrexia with no chorioamnionitis: Pyrexia with chorioamnionitis: National Maternity Network:
(suspected sepsis) Gram-negative organisms Cephalosporin with activity add metronidazole. Management of Intrapartum Maternal
Anaerobes against GBS (e.g., cefotaxime) Pyrexia in Hospital Guideline. NHS
Penicillin allergy: Vancomycin Scotland. Scottish Perinatal Network.
2022 [ 145 ]
Mastitis causing sepsis MRSA Flucloxacillin + clindamycin Confirmed MRSA: Vancomycin Bacterial Sepsis in Pregnancy.
Streptococci Penicillin allergy: Green–top Guideline No. 64a. RCOG
Vancomycin + clindamycin or 2012 [ 146 ]
Clindamycin/teicoplanin Mastitis and breast abscess. BMJ Best
Practise. BMJ 2023 [ 147 ]
Caesarean section wound MRSA Flucloxacillin + clindamycin Bacterial Sepsis in Pregnancy.
infection causing sepsis Streptococci Penicillin allergy: Green–top Guideline No. 64a. RCOG
Vancomycin + clindamycin or 2012 [ 146 ]
clindamycin/teicoplanin
Endometritis causing Gram-negative anaerobes Gentamicin one dose Bacterial Sepsis in Pregnancy.
sepsis Streptococci immediately + cefo- Green–top Guideline No. 64a. RCOG
taxime + metronidazole or 2012 [ 146 ]
gentamicin + clindamycin Antibiotic regimens for postpartum
Penicillin allergy: endometritis. Cochrane Database Syst
Gentamicin + clin- Rev. 2015 [ 148 ]
damycin + ciprofloxacin
Acute pyelonephritis Gram-negative bacteria Gentamicin (once ESBLs: gentamicin + meropenem Bacterial Sepsis in Pregnancy.
causing sepsis Staphylococci and only) + cefotaxime or just Green–top Guideline No. 64a. RCOG
Streptococci cefuroxime 2012 [ 146 ]
Penicillin allergy: Pyelonephritis (acute): antimicrobial
Gentamicin + ciprofloxacin prescribing. NICE guideline [NG111].
NICE 2018 [ 149 ]
Severe sepsis (with no MRSA, streptococci, Gentamicin (once Bacterial Sepsis in Pregnancy.
clear focus) Gram-negatives (including only) + meropenem + clin- Green–top Guideline No. 64a. RCOG
ESBL damycin 2012 [ 146 ]
producers + Pseudomonas) Penicillin allergy:
and anaerobes Clindamycin + gentamicin +
metronidazole + ciprofloxacin
Toxic shock syndrome Staphylococci Streptococci Gentamicin (once only) + flu- Confirmed Strep: benzylpenicillin Bacterial Sepsis in Pregnancy.
cloxacillin + clindamycin or and clindamycin. Green–top Guideline No. 64a. RCOG
carbapenem (i.e., Confirmed methicillin sensitive 2012 [ 146 ]
imipenem/cilastatin or Staph: Toxic shock syndrome. BMJ Best
meropenem); a penicillin with a clindamycin + oxacillin/nafcillin. Practise. BMJ 2023 [ 150 ]
beta-lactamase inhibitor (e.g., Confirmed MRSA: vancomycin
ticarcillin/clavulanate or instead of flucloxacillin. Regimen
piperacillin/tazobactam); must contain an antitoxin agent
Penicillin allergy: such as clindamycin or linezolid.
Gentamicin (once only) + van- Also Consider IVIG
comycin + clindamycin or
gentamicin (once
only) + linezolid or just
vancomycin
(continued on next page)

7
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

Table 1 (continued)

Indication Causative organisms Antibiotic recommendations Important notes References

Neonatal (EOS)

Early-onset neonatal Streptococcus (GBS); Gram- Benzylpenicillin with gentamicin Neonatal infection: antibiotics for
sepsis negative bacteria prevention and treatment NICE
guideline [NG195]. NICE 2021 [ 151 ]
ACOG: American College of Obstetricians and Gynecologists; EOS: Early onset neonatal sepsis; ESBLs: Extended-spectrum beta-lactamases; GBS: Group B Strepto-
coccus; IVIG: Intravenous immunoglobulin; MRSA: Methicillin-resistant Staphylococcus aureus; NICE: National Institute for Health and Care Excellence; RCOG: Royal
College of Obstetricians & Gynaecologists; WHO: World Health Organization.

are much higher. For example, in Sudan up to 92% of urinary needed to establish means of surveillance for effective steward-
E. coli isolates are resistant.[ 154 ] For Sudanese healthcare, the ship programs in vulnerable neonatal populations in different
key drivers of AMR include widespread inappropriate antibiotic healthcare settings.
use with up to 65% of hospitalized patients receiving antibi-
otics, and poor access to microbial sensitivity results to guide Types of antimicrobial stewardship interventions
better prescribing.[ 155–157 ] In these resource-limited countries,
much of the inappropriate use is due to empirical treatment or The WHO has published a framework of antimicrobial stew-
the use of antibiotic prophylaxis.[ 158 ] AMR is estimated to re- ardship interventions for LMIC, but these are also relevant for
sult in 700,000 deaths each year globally. In 2015 the WHO en- resource-rich countries.[ 167 ] Similarly, NICE has published guid-
dorsed a Global Action Plan on Antimicrobial Resistance (GAP) ance for the UK.[ 168 ] The interventions are centered around im-
to address the global problem, and in the UK a government doc- proved education, multidisciplinary working, and better pre-
ument was published in 2019 with the country’s 5-year action scribing. In other patient groups, this multifaceted approach has
plan, which amongst other goals, was aimed at reducing antibi- been shown to reduce antibiotic prescriptions without increas-
otic use by 15% and specified drug-resistant infections by 10% ing the risk of complications.[ 169 ] In addition, appropriate an-
over 5 years.[ 159 ] In the maternity setting, patients with peripar- tibiotic use can reduce the rates of hospital acquired infections
tum infection have been shown to have resistance to a number such as Clostridium difficile.[ 170 ]
of common organisms encountered in obstetrics such as E. coli, In brief, the proposed recommended interventions comprise
[ 167–169 ]
GBS and GAS, with resistance rates as high as 62% to 81% for : (1) basic resources to support antibiotic use including
E. coli.[ 160 ] Cohort-specific antibiograms with isolates and their local guidelines for treatment and prophylaxis, access to a mi-
sensitivities may provide a better way to determine resistance crobiology laboratory and clinical expertise on infections, phar-
patterns to guide antibiotic prescribing in maternity.[ 161 ] macy overview of dosing and duration of treatments. (2) tools
such as the quick sequential (sepsis-related) organ failure as-
Healthcare associated infections sessment (qSOFA) tool for pregnant women, and the Kaiser Per-
manente sepsis risk calculator for neonates can enable better
The other consequence of widespread antibiotic use is the recognition of sepsis and improve decision-making surrounding
development of HCAIs. The HCAI rate in the UK and devel- the need for antibiotics. (3) education of healthcare profession-
oped countries ranges between 3.5% and 12%. In the inten- als to understand indications for treatments, when treatments
sive care unit (ICU) up to 30% of patients are affected by at should be reviewed, use of decision tools, and the importance of
least one episode of HCAI. In comparison, in LMIC the incidence auditing and recording antibiotic use, prescribing, and clinical
is greater, averaging between 5.7% and 19.1%, and ICU infec- outcomes.(4) use of resources to improve prescribing and effec-
tions could be as high as 88%.[ 162 ] Poor reporting and lack of tiveness of antimicrobial stewardship that includes local and re-
surveillance schemes mean the true incidence may be higher. gional surveillance of AMR (in common organisms/infections),
In the UK, annual counts of hospital-acquired Clostridioides dif- rapid testing (such as rapid polymerase chain reaction [PCR] as
ficile and Methicillin-resistant Staphylococcus aureus (MRSA) in- discussed later) and restrictive prescribing of certain antibiotics
fections have been falling since 2007 and are currently show- to prevent resistance.
ing a 6 and 7-fold decrease respectively when compared to
2007.[ 163 , 164 ] However, these rates have remained unchanged What Research Gaps Need to be Addressed to Further
since 2013 and new efforts are required to ensure a continued Improve Clinical Management and Treatment of Maternal
decrease. Sepsis?

The need for neonatal antimicrobial stewardship Whilst the overall UK prevalence of sepsis-related maternal
deaths remains low, sepsis is still one of the leading causes of di-
Antibiotic prescribing in pediatric populations also remains rect death in this population. Since 2011, efforts have been made
a concern and an area that requires attention. The WHO Access, to highlight the importance of better clinical management of
Watch, and Reserve classification was used in a study across 56 sepsis in maternal clinical pathways with early recognition and
countries to assess the patterns of antibiotic use.[ 165 ] The study treatment.[ 146 , 171 ] These have included recommendations from
identified significant variation in antibiotic use in hospitalized Mothers and Babies: Reducing Risk through Audits and Confi-
neonates and children. One of the key barriers to effective stew- dential Enquiries (MBRRACE), NICE and Royal College of Obste-
ardship in neonatal populations remains the lack of metrics for tricians & Gynaecologists (RCOG) guidelines as well as screen-
measuring the quality of antibiotic use.[ 166 ] Much more effort is ing and action tools from the UK Sepsis trust.[ 138 , 146 , 171 , 172 ]

8
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

Furthermore, initiatives such as the maternity early warning Immunotherapy agents


score (MEWS) and sepsis six have been shown to effectively The RECOVERY trial collaborative’s subsequent work in-
predict morbidity in maternity patients and reduce inpatient vestigating the benefits of Tocilizumab treatment, which is a
mortality respectively.[ 137 , 173 , 174 ] This has been relatively effec- monoclonal antibody targeted against the interleukin-6 recep-
tive with a reduction in direct deaths due to sepsis in the UK tor, found that in hospitalized COVID-19 patients with hypoxia
from 0.63 to 0.44 per 100,000 births from 2009–11 to 2015–17 and systemic inflammation, Tocilizumab not only improved sur-
respectively.[ 175 ] Alongside these, rates of EOS have also been vival, but it also reduced the likelihood of the composite end-
falling.[ 176 , 177 ] However, many maternal deaths remain poten- point of invasive mechanical ventilation or death.[ 185 ]
tially preventable. To date, randomized controlled trials conducted using
immune-modulators such as Granulocyte-macrophage colony-
Tools to improve recognition of maternal sepsis stimulating factor (GM-CSF) and IgM-enriched immunoglob-
ulin have shown promising results with improved resolution
Better recognition and prophylactic treatment may provide from infection and reduced mortality risk respectively.[ 186 , 187 ]
useful methods to prevent sepsis, particularly during the peri- Other potential targets for improved host responses include
partum period. Similar to SRC, obstetric-modified sepsis scoring IFN-𝛾, IL-7, and IL-15, which have been investigated ex vivo,
tools are useful to objectively risk stratify the severity of sepsis with evidence showing that their use results in improved leu-
and direct clinical treatment.[ 178 ] This includes the qSOFA tool cocyte responses.[ 188–191 ] Other agents are an IL-1R antagonist
for identifying critically ill obstetric patients.[ 178 , 179 ] (Anakinra), shown to be safe during pregnancy in small studies
and used in sepsis in non-pregnant patients (macrophage ac-
tivating syndrome) to reduce mortality.[ 192 , 193 ] Unfortunately,
Interventions as prophylaxis comparing previous clinical studies to make clear recommen-
dations is difficult due to their heterogeneous study designs
For prophylaxis, the prophylactic antibiotics in the preven- with myriad differences including treatment dosing, inclusion
tion of infection after operative vaginal delivery (ANODE) study and exclusion criteria, supportive treatments, settings, and en-
and recent Cochrane review on vaginal antiseptic preparation listed study populations. Furthermore, circulating cytokine lev-
prior to Caesarean section found that such interventions resulted els vary during severe sepsis meaning that clinical benefit may
in a 40% reduction in suspected or confirmed infection, and a only be seen in some patients and suggesting that their use
64% reduction in post‐Caesarean endometritis respectively with should be reserved for certain situations.[ 194 ] Indeed, anti-TNF-𝛼
likely limited impact on AMR.[ 20 , 180 ] In LMIC, implementing immunotherapy is associated with an overall reduction in mor-
these as part of a series of interventions to reduce maternal and tality and improved survival in patients with high serum lev-
neonatal sepsis would be required in order to be effective.[ 181 ] els of IL-6.[ 195 ] Observational data from patients taking disease
modifying drugs for immune-mediated medical conditions such
Immunomodulators as rheumatoid arthritis suggest immune suppression may help
to prevent unregulated host responses to sepsis.[ 196 ]
In addition to antibiotics, research to find new methods to Clearly in current times with emerging viral pandemics, a
enhance either the acute response to infection or the recovery significant global incidence of bacterial sepsis, high AMR, and
phase during sepsis are potentially important areas for future the need for better antibiotic stewardship, novel methods of reg-
work. Poor acute immune responses have been associated with ulating host responses to infection are an important area of re-
an increased risk of developing secondary infections, whilst ex- search. A good example is the GBS vaccine, which has thus far
cessive inflammatory responses are associated with an increased been investigated in early clinical trials in pregnancy and shows
risk of developing downstream complications from sepsis such a great deal of promise.[ 197 ] Furthermore, the development of
as cardiovascular collapse.[ 182 , 183 ] such a vaccine has been identified as a research priority by the
WHO.
Steroid immune suppression
Respiratory complications during sepsis are likely to ben- What is the Evidence for Personalised Approaches to Treat
efit most from immunosuppression. Animal study data sug- Maternal Infections?
gests that this may be particularly relevant in pregnancy. A
more recent example of a clinical trial of immunomodulators Preterm birth and chorioamnionitis provide an area of clini-
to modify the host response is the novel Randomised Evalua- cal research where a personalized approach to the management
tion of COVID-19 Therapy (RECOVERY Trial) comparing dif- of infection may reduce the risk of maternal sepsis as well as im-
ferent treatments aimed at altering the host response to SARS- prove neonatal outcomes. In fact, infection-driven preterm birth
CoV-2 virus (NCT04381936). In patients recruited during the is one of the only aetiologies that has a proven direct causal link.
first wave of the pandemic between March and June 2020, dex-
amethasone treatment significantly lowered 28-day mortality in Preterm birth as a model for obstetric infections
patients receiving either invasive mechanical ventilation or oxy-
gen alone.[ 184 ] Importantly and unlike many other clinical tri- It is increasingly perceived that the uterine cavity and amni-
als, RECOVERY explicitly included pregnant women during re- otic fluid are not sterile, but that they both contain a rich micro-
cruitment. However, despite showing a clear clinical benefit, far biome that is different in each compartment.[ 198 ] The uterine mi-
fewer critically ill pregnant women compared to non-pregnant crobiome appears synchronous with vaginal bacterial colonies
women received steroids (9.3% vs. 22.6%). and is affected by age, endometrial inflammation (endometri-

9
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

tis), and mode of birth (cesarean section or vaginal birth)[ 199 ] More recently, in a non-human primate model of preterm
whereas the amniotic cavity appears to share its constituent mi- birth using an intrauterine injection of E. coli, administration
crobiota with the fetus, exposing it to microbes throughout its of systemic antibiotics 24 h after injecting E. coli was signifi-
in-utero development probably by ingestion.[ 200 , 201 ] In almost cantly effective at eradicating maternal bacteremia. However,
60% of cases of PTL is thought to be due to microbial invasion treatment did not resolve choriodecidual or amnion tissue in-
of the amniotic cavity (MIAC). These invading organisms will of- flammation nor preterm birth.[ 211 ] These studies suggest that
ten include those linked with preterm birth such as Mycoplasma antibiotic prophylaxis, and appropriate treatment of chorioam-
and Ureaplasma species,[ 202 , 203 ] which are typically not present nionitis can prevent maternal sepsis but they may not improve
in mid-trimester pregnancies.[ 204 ] fetal or neonatal outcomes. So, how can clinical teams rapidly
detect bacterial infections and target antibiotic treatments?

Antibiotics to treat intra-amniotic infection New methods to detect pathogens

Professor Sara Kenyon and the ORACLE collaborative group Whereas traditional microscopy and culture-based methods
in the early 2000′s designed a randomized controlled trial of have often described sterile or culture-negative inflammation in
erythromycin or co-amoxiclav given to pregnant women pre- amniotic fluid with preterm birth, new techniques using nucleic
senting with PTL and/or preterm ruptured membranes. Impor- acid amplification testing (NAAT) or amplification of prokary-
tantly, they showed treatment resulted in a significant reduction otic 16S subunit ribosomal DNA have proven to be better de-
in maternal infection.[ 205 ] Their findings also demonstrated a tection tools, identifying a broad range of organisms as well as
short-term improvement in neonatal morbidity, but no reduc- those that prove to be more difficult to detect using standard
tion in perinatal mortality or improvement in the health of the culture-based methods such as Ureaplasma.[ 212 ]
children at age seven.[ 206–208 ] However, when administered to One of the drawbacks of 16S is the speed of the assay since
the intact membrane group in PTL, treatment with either an- a reference laboratory is usually required meaning that results
tibiotic did not improve neonatal outcomes.[ 209 , 210 ] can take several days from sample collection. Furthermore, dif-

Figure 2. Use of technologies to detect systemic and intra-amniotic infection. Ascending organisms MIAC into the amniotic cavity will be phagocytosed by neu-
trophils, PAMPs originating from these organisms will activate macrophages causing these cells to release pro-inflammatory cytokines (such as IL-6). IL-6 is also
produced by activated monocytes, and amnion cells along with other reproductive/pregnancy tissue as part of an inflammatory immune response. Amniotic fluid
can be sampled (by amniocentesis) and used to detect both intra-amniotic infection and inflammation. Systemic infection can be detected from peripheral blood.
Traditional NAAT methods can be relatively rapid and are highly sensitive but lack the isolation of AMR. 16S rRNA methods are an alternative, metagenomics
gives better coverage, and new methods such as multiplex PCR are rapid and suitable as a POC. Inflammation can be measured by ELISA, rapid chemiluminescent
immunoassay and by a lateral flow POC.
AMR: Antimicrobial resistance; ELISA: Enzyme linked immunosorbent assay; IL-6: Interleukin-6; MIAC: Microbial invasion of the amniotic cavity; NAAT: Nucleic
acid amplification test; PAMPs: Pathogen associated molecular patterns; PCR: Polymerase chain reaction; POC: Point-of-care test; rRNA: ribosomal RNA.

10
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

ferentiating between commensals and colonizing organisms can elevated levels of IL-6 and labeled these as infective pathogens.
be difficult. By comparison, culture and sensitivities may take Concentrations of IL-6 in the fluid were combined with the intra-
48 h or more but provide more clinically useful information. amniotic micro-organisms identified, to create a multivariate
New techniques that can identify organisms quickly, and those regression model for predictors of latency to birth, composite
that also sequence any AMR genes have begun to emerge, which perinatal morbidity, and mortality rates, adjusted for gestational
provide a more comprehensive output for clinical application. age. Their results showed that IL-6 was a better predictor of poor
For example, in the context of labor and birth, NAAT of GBS pro- outcomes than the microbial species alone, which was in keep-
vides rapid diagnosis with good sensitivity and specificity (91% ing with previous reports.[ 216 , 217 ]
and 98% respectively) to enable more accurate management of Equally, the Premature Rupture Membranes and threatened
laboring mothers.[ 213 ] Methods such as nanopore metagenomic PTL - a Personalised Approach (PROMPT) will investigate the
sequencing can genotype without the need for amplification or clinical benefits of rDNA methods using a multiplex-based PCR
labeling. Proof of concept studies have demonstrated the fea- system to detect bacteria and cytokine profiling of amniotic
sibility of nanopore technology in respiratory and blood sam- fluid with the aim of improving latency to birth as well as ma-
ples with good concordance with traditional culture and sensi- ternal infection rates in women with ruptured membranes and
tivity, thus providing clinical teams the ability to identify or- PTL (Figure 2).[ 218 ] Indeed, the use of omics, whereby molecule
ganisms, and resistance patterns and rapidly direct appropriate groups can be assessed from the same biological sample, may
anti-microbial treatments.[ 214 , 215 ] provide a comprehensive look at the pathophysiology of the pro-
cess in question such as with sepsis.[ 219 ]
Improving the precision of identifying clinically relevant
infections Conclusions

However, with early detection at presentation and prior to The key strategies discussed in this manuscript are sum-
pregnant women developing overt clinical signs of severe in- marised in Figure 3. Maternal sepsis remains a global threat
fection, can this technology be further refined to establish a to safe pregnancy, childbirth, and maternal and neonatal out-
likelihood of severity? Here, cytokine profiling of reservoirs of comes. In the UK, sepsis rates have steadily been falling and
organisms such as the amniotic cavity may be helpful. Combs emerging research such as the ANODE study and Cochrane re-
et al.[ 202 ] investigated amniotic fluid infection in women with view on pre-Caesarean section vaginal preparation will reduce
spontaneous PTL with intact fetal membranes using amniocen- these rates further. Despite this, sepsis remains an important
tesis alongside standard culture and 16S rDNA methods. They cause of maternal morbidity and mortality in the UK. Initiatives
correlated the most prevalent organisms in amniotic fluid with such as the global maternal sepsis study (GLOSS) and NeoAMR,

Figure 3. Key strategies for the management of sepsis in pregnancy. This flowchart summaries the maternal neonatal complications, the importance of prompt
antibiotic use but the potential issues with the current usage, how antibiotic stewardship can be achieved and the potential for adjunctive treatments.
AMR: Antimicrobial resistance; ANODE: Prophylactic antibiotics in the prevention of infection after operative vaginal delivery; GLOSS: Global maternal sepsis study;
MBRRACE: Mothers and Babies: Reducing Risk through Audits and Confidential Enquiries; NAAT: Nucleic acid amplification test; PCR: Polymerase chain reaction;
qSOFA: quick Sequential organ failure assessment; rDNA: ribosomal DNA; SRC: Sepsis risk calculator.

11
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

will help to provide a framework for reporting maternal infec- References


tions as well as reflecting the burden of neonatal unit-level AMR
[1] Fleischmann C, Scherag A, Adhikari NK, Hartog CS, Tsaganos T, Schlattmann P,
worldwide. This will guide the development of core practices for et al. Assessment of global incidence and mortality of hospital-treated sepsis. Cur-
managing maternal and neonatal infections. Early and appropri- rent estimates and limitations. Am J Respir Crit Care Med 2016;193(3):259–72.
doi:10.1164/rccm.201504-0781OC.
ate detection of both maternal and neonatal infections is key and [2] World Health Organization [Internet]. World Health Organisation: health top-
tools such as qSOFA and SRC have the potential to influence an- ics/fact sheets/ sepsis. Available from: https://www.who.int/news-room/fact-
tibiotic stewardship and thus clinical care in the acute setting. sheets/detail/sepsis. [Accessed 19th April 2018].
[3] Say L, Chou D, Gemmill A, Tunçalp Ö, Moller AB, Daniels J, et al. Global causes of
However, further research is required on their use. Adjunctive maternal death: a WHO systematic analysis. Lancet Glob Health 2014;2(6):e323–
immunotherapy is another potential avenue for research in sep- 33. doi:10.1016/S2214-109X(14)70227-X.
[4] Black RE, Levin C, Walker N, Chou D, Liu L, Temmerman M. Repro-
sis whereby the host response can be altered to fit the patient’s ductive, maternal, newborn, and child health: key messages from Dis-
needs in terms of both clinical deterioration and immune re- ease Control Priorities 3rd Edition. Lancet 2016;388(10061):2811–24.
doi:10.1016/S0140-6736(16)00738-8.
sponse. This introduces the concept of personalized medicine [5] Chen L, Wang Q, Gao Y, Zhang J, Cheng S, Chen H, et al. The global bur-
where combinations of antibiotics and immunotherapy agents den and trends of maternal sepsis and other maternal infections in 204 coun-
are used together to improve the clinical outcome. Individual- tries and territories from 1990 to 2019. BMC Infect Dis 2021;21(1):1074.
doi:10.1186/s12879-021-06779-0.
ized risk stratification rather than the use of population-based [6] Bonet M, Nogueira Pileggi V, Rijken MJ, Coomarasamy A, Lissauer D,
scores and protocols will help to achieve this. There is still some Souza JP, et al. Towards a consensus definition of maternal sepsis: results of
a systematic review and expert consultation. Reprod Health 2017;14(1):67.
way to go before this is a clinically viable option but research to doi:10.1186/s12978-017-0321-6.
date has yielded promising results. It remains critical in the cur- [7] Acosta CD, Kurinczuk JJ, Lucas DN, Tuffnell DJ, Sellers S, Knight M. Severe
maternal sepsis in the UK, 2011-2012: a national case-control study. PLoS Med
rent climate where patients often face multi-organism infection 2014;11(7):e1001672. doi:10.1371/journal.pmed.1001672.
and widespread antibiotic drug resistance, to investigate novel [8] Acosta CD, Harrison DA, Rowan K, Lucas DN, Kurinczuk JJ, Knight M. Maternal
morbidity and mortality from severe sepsis: a national cohort study. BMJ Open
methods to diagnose and manage sepsis. 2016;6(8):e012323. doi:10.1136/bmjopen-2016-012323.
[9] Kidson KM, Henderson WR, Hutcheon JA. Case fatality and adverse outcomes
are reduced in pregnant women with severe sepsis or septic shock com-
Author Contributions
pared with age-matched comorbid-matched nonpregnant women. Crit Care Med
2018;46(11):1775–82. doi:10.1097/CCM.0000000000003348.
NMS was responsible for the conception of the article topic, [10] Haddad LB, Horton J, Ribner BS, Jamieson DJ. Ebola infection in pregnancy: a
global perspective and lessons learned. Clin Obstet Gynecol 2018;61(1):186–96.
undertook the primary literature search, reviewed, and sum- doi:10.1097/GRF.0000000000000332.
marised current evidence, was responsible for writing the ini- [11] Mosby LG, Rasmussen AS, Jamieson JD. 2009 pandemic influenza A (H1N1)
in pregnancy: a systematic review of the literature. Am J Obstet Gynecol
tial and revised drafts based on comments and suggestions from 2011;205(1):10–18. doi:10.1016/j.ajog.2010.12.033.
co‐authors and submitted the final version. EC, DM, and FCC [12] Wong SF, Chow KM, Leung TN, Ng WF, Ng TK, Shek CC, et al. Pregnancy and
perinatal outcomes of women with severe acute respiratory syndrome. Am J Obstet
all reviewed each draft with critical revision of intellectual con- Gynecol 2004;191(1):292–7. doi:10.1016/j.ajog.2003.11.019.
tent, reviewed the current literature and incorporated this into [13] Di Mascio D, Khalil A, Saccone G, Rizzo G, Buca D, Liberati M, et al. Outcome of
the article. MRJ was involved in the conception of the article coronavirus spectrum infections (SARS, MERS, COVID-19) during pregnancy: a sys-
tematic review and meta-analysis. Am J Obstet Gynecol MFM 2020;2(2):100107.
topic, critically revised the intellectual content, and advised on doi:10.1016/j.ajogmf.2020.100107.
the structure of the article, content, and current areas of debate. [14] Schwartz DA, Graham AL. Potential maternal and infant outcomes from
(Wuhan) coronavirus 2019-nCoV infecting pregnant women: lessons from SARS,
All authors read and approved the final version. MERS, and other human coronavirus infections. Viruses 2020;12(2):194.
doi:10.3390/v12020194.
[15] Tang P, Wang J, Song Y. Characteristics and pregnancy outcomes of patients
Acknowledgements with severe pneumonia complicating pregnancy: a retrospective study of 12
cases and a literature review. BMC Pregnancy Childbirth 2018;18(1):434.
doi:10.1186/s12884-018-2070-0.
Images used in the figures were created with Biorender.com. [16] Blauvelt CA, Nguyen KC, Cassidy AG, Gaw SL. Perinatal outcomes among pa-
tients with sepsis during pregnancy. JAMA Netw Open 2021;4(9):e2124109.
doi:10.1001/jamanetworkopen.2021.24109.
Funding [17] Renaud SJ, Cotechini T, Quirt JS, Macdonald-Goodfellow SK, Othman M, Gra-
ham CH. Spontaneous pregnancy loss mediated by abnormal maternal in-
This research did not receive any specific grant from funding flammation in rats is linked to deficient uteroplacental perfusion. J Immunol
2011;186(3):1799–808. doi:10.4049/jimmunol.1002679.
agencies in the public, commercial, or not-for-profit sectors. [18] The WHO Global Maternal Sepsis Study (GLOSS) Research GroupFrequency and
management of maternal infection in health facilities in 52 countries (GLOSS):
a 1-week inception cohort study. Lancet Glob Health 2020;8(5):e661–ee71.
Ethics Statement doi:10.1016/S2214-109X(20)30109-1.
[19] Knowles SJ, O’Sullivan NP, Meenan AM, Hanniffy R, Robson M. Maternal sepsis
incidence, aetiology and outcome for mother and fetus: a prospective study. BJOG
Not applicable. 2015;122(5):663–71. doi:10.1111/1471-0528.12892.
[20] Knight M, Chiocchia V, Partlett C, Rivero-Arias O, Hua X, Hinshaw K, et al. Prophy-
lactic antibiotics in the prevention of infection after operative vaginal delivery (AN-
Conflict of Interest ODE): a multicentre randomised controlled trial. Lancet 2019;393(10189):2395–
403. doi:10.1016/S0140-6736(19)30773-1.
[21] Cornelissen L, Woodd S, Shakur-Still H, Fawole B, Noor S, Etuk S, et al. Sec-
The authors declare that they have no known competing fi- ondary analysis of the WOMAN trial to explore the risk of sepsis after invasive
nancial interests or personal relationships that could have ap- treatments for postpartum hemorrhage. Int J Gynaecol Obstet 2019;146(2):231–
peared to influence the work reported in this paper. 7. doi:10.1002/ijgo.12860.
[22] Gentilotti E, De Nardo P, Nguhuni B, Piscini A, Damian C, Vairo F, et al. Implement-
ing a combined infection prevention and control with antimicrobial stewardship
Data Availability joint program to prevent caesarean section surgical site infections and antimicro-
bial resistance: a Tanzanian tertiary hospital experience. Antimicrob Resist Infect
Control 2020;9(1):69. doi:10.1186/s13756-020-00740-7.
The data sets generated during and/or analyzed during the [23] Weinberg M, Fuentes JM, Ruiz AI, Lozano FW, Angel E, Gaitan H, et al. Reducing
infections among women undergoing cesarean section in Colombia by means of
current study are available from the corresponding author upon continuous quality improvement methods. Arch Intern Med 2001;161(19):2357–
reasonable request. 65. doi:10.1001/archinte.161.19.2357.

12
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

[24] Zöllner J, Howe LG, Edey LF, O’Dea KP, Takata M, Gordon F, et al. The response of [50] Szekeres-Bartho J, Faust Z, Varga P. The expression of a progesterone-induced
the innate immune and cardiovascular systems to LPS in pregnant and nonpregnant immunomodulatory protein in pregnancy lymphocytes. Am J Reprod Immunol
mice. Biol Reprod 2017;97(2):258–72. doi:10.1093/biolre/iox076. 1995;34(6):342–8. doi:10.1111/j.1600-0897.1995.tb00962.x.
[25] Zöllner J, Lambden S, Nasri NM, Leiper J, Johnson MR. Rapid onset of severe septic [51] Somerset DA, Zheng Y, Kilby MD, Sansom DM, Drayson MT. Normal hu-
shock in the pregnant mouse†. Biol Reprod 2019;100(2):505–13. doi:10.1093/bi- man pregnancy is associated with an elevation in the immune suppres-
olre/ioy193. sive CD25+ CD4+ regulatory T-cell subset. Immunology 2004;112(1):38–43.
[26] Zölner J, Lambden S, Nasri NM, Johnson MR, Leiper J. Inhibition of Dimethylargi- doi:10.1111/j.1365-2567.2004.01869.x.
nine Dimethylaminohydrolase 1 Improves the Outcome of Sepsis in Pregnant Mice. [52] Shigeta N, Kumasawa K, Tanaka A, Badger Wing J, Nakamura H, Sakaguchi S, et al.
Shock 2020;54(4):498–506. doi:10.1097/SHK.0000000000001490. Dynamics of effector and naïve regulatory T cells throughout pregnancy. J Reprod
[27] Aronoff DM, Mulla ZD. Postpartum invasive group A streptococcal dis- Immunol 2020;140:103135. doi:10.1016/j.jri.2020.103135.
ease in the modern era. Infect Dis Obstet Gynecol 2008;2008:796892. [53] Bulmer JN, Williams PJ, Lash GE. Immune cells in the placental bed. Int J Dev Biol
doi:10.1155/2008/796892. 2010;54(2–3):281–94. doi:10.1387/ijdb.082763jb.
[28] Palaniappan N, Menezes M, Willson P. Group A streptococcal puerperal [54] Lash GE, Robson SC, Bulmer JN. Review: functional role of uterine natural killer
sepsis: management and prevention. Obstet Gynaecol 2012;14(1):9–16. (uNK) cells in human early pregnancy decidua. Placenta 2010(31 Suppl):S87–92.
doi:10.1111/j.1744-4667.2011.00082.x. doi:10.1016/j.placenta.2009.12.022.
[29] Deutscher M, Lewis M, Zell ER, Taylor TH Jr, Van Beneden C, Schrag S. Incidence [55] Vento-Tormo R, Efremova M, Botting RA, Turco MY, Vento-Tormo M, Meyer KB,
and severity of invasive Streptococcus pneumoniae, group A Streptococcus, and et al. Single-cell reconstruction of the early maternal-fetal interface in humans.
group B Streptococcus infections among pregnant and postpartum women. Clin Nature 2018;563(7731):347–53. doi:10.1038/s41586-018-0698-6.
Infect Dis 2011;53(2):114–23. doi:10.1093/cid/cir325. [56] Pique-Regi R, Romero R, Tarca AL, Sendler ED, Xu Y, Garcia-Flores V, et al. Single
[30] Leonard A, Wright A, Saavedra-Campos M, Lamagni T, Cordery R, Nicholls M, et al. cell transcriptional signatures of the human placenta in term and preterm parturi-
Severe group A streptococcal infections in mothers and their newborns in London tion. eLife 2019;8:e52004. doi:10.7554/eLife.52004.
and the South East, 2010-2016: assessment of risk and audit of public health man- [57] Strunz B, Bister J, Jönsson H, Filipovic I, Crona-Guterstam Y, Kvedaraite E, et al.
agement. BJOG 2019;126(1):44–53. doi:10.1111/1471-0528.15415. Continuous human uterine NK cell differentiation in response to endometrial re-
[31] Billington WD. The immunological problem of pregnancy: 50 years with the hope generation and pregnancy. Sci Immunol 2021;6(56):eabb7800. doi:10.1126/sci-
of progress. A tribute to Peter Medawar. J Reprod Immunol 2003;60(1):1–11. immunol.abb7800.
doi:10.1016/s0165-0378(03)00083-4. [58] Xu Y, Romero R, Miller D, Silva P, Panaitescu B, Theis KR, et al. Innate lymphoid
[32] Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of foreign cells. cells at the human maternal-fetal interface in spontaneous preterm labor. Am J
Nature 1953;172(4379):603–6. doi:10.1038/172603a0. Reprod Immunol 2018;79(6):e12820. doi:10.1111/aji.12820.
[33] Medawar P. Some immunological and endocrinological problems raised by the evo- [59] Mendes J, Rodrigues-Santos P, Areia AL, Almeida J-S, Alves V, Santos-Rosa M, et al.
lution of viviparity. Symposia of the society for experimental biology. Cambridge: Type 2 and type 3 innate lymphoid cells at the maternal-fetal interface: implications
Cambridge University Press; 1953. in preterm birth. BMC Immunol 2021;22(1):28. doi:10.1186/s12865-021-00423-x.
[34] Mor G, Cardenas I. The immune system in pregnancy: a unique complexity. Am J [60] Houser BL. Decidual macrophages and their roles at the maternal-fetal interface.
Reprod Immunol 2010;63(6):425–33. doi:10.1111/j.1600-0897.2010.00836.x. Yale J Biol Med 2012;85(1):105–18.
[35] Shah NM, Herasimtschuk AA, Boasso A, Benlahrech A, Fuchs D, Imami N, et al. [61] Sayama S, Nagamatsu T, Schust DJ, Itaoka N, Ichikawa M, Kawana K, et al. Human
Changes in T cell and dendritic cell phenotype from mid to late pregnancy are decidual macrophages suppress IFN-𝛾 production by T cells through costimulatory
indicative of a shift from immune tolerance to immune activation. Front Immunol B7-H1:PD-1 signaling in early pregnancy. J Reprod Immunol 2013;100(2):109–17.
2017;8:1138. doi:10.3389/fimmu.2017.01138. doi:10.1016/j.jri.2013.08.001.
[36] Shah NM, Imami N, Johnson MR. Progesterone modulation of pregnancy-related [62] Wang SC, Li YH, Piao HL, Hong XW, Zhang D, Xu YY, et al. PD-1 and Tim-3 path-
immune responses. Front Immunol 2018;9:1293. doi:10.3389/fimmu.2018. ways are associated with regulatory CD8+ T-cell function in decidua and main-
01293. tenance of normal pregnancy. Cell Death Dis 2015;6(5):e1738. doi:10.1038/cd-
[37] Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that dis.2015.112.
sustains anergy to fetal antigen. Nature 2012;490(7418):102–6. doi:10.1038/na- [63] Tilburgs T, Schonkeren D, Eikmans M, Nagtzaam NM, Datema G, Swings GM,
ture11462. et al. Human decidual tissue contains differentiated CD8+ effector-memory T
[38] Schober L, Radnai D, Schmitt E, Mahnke K, Sohn C, Steinborn A. Term and preterm cells with unique properties. J Immunol 2010;185(7):4470–7. doi:10.4049/jim-
labor: decreased suppressive activity and changes in composition of the regulatory munol.0903597.
T-cell pool. Immunol Cell Biol 2012;90(10):935–44. doi:10.1038/icb.2012.33. [64] Kahn DA, Baltimore D. Pregnancy induces a fetal antigen-specific maternal T
[39] Shah NM, Edey LF, Imami N, Johnson MR. Human labour is associated with al- regulatory cell response that contributes to tolerance. P Natl Acad Sci USA
tered regulatory T cell function and maternal immune activation. Clin Exp Immunol 2010;107(20):9299–304. doi:10.1073/pnas.1003909107.
2020;199(2):182–200. doi:10.1111/cei.13384. [65] Benner M, Feyaerts D, García CC, Inci N, López SC, Fasse E, et al. Clusters of
[40] Taglauer ES, Yankee TM, Petroff MG. Maternal PD-1 regulates accumulation of tolerogenic B cells feature in the dynamic immunological landscape of the preg-
fetal antigen-specific CD8+ T cells in pregnancy. J Reprod Immunol 2009;80(1– nant uterus. Cell Rep 2020;32(13):108204. doi:10.1016/j.celrep.2020.108204.
2):12–21 S0165-0378(09)00038-2 [pii] . doi:10.1016/j.jri.2008.12.001. [66] Forbes RL, Gibson PG, Murphy VE, Wark PA. Impaired type I and III inter-
[41] Lissauer D, Piper K, Goodyear O, Kilby MD, Moss PA. Fetal-specific CD8+ cy- feron response to rhinovirus infection during pregnancy and asthma. Thorax
totoxic T cell responses develop during normal human pregnancy and exhibit 2012;67(3):209–14. doi:10.1136/thoraxjnl-2011-200708.
broad functional capacity. J Immunol 2012;189(2):1072–80. doi:10.4049/jim- [67] Shah NM, Imami N, Kelleher P, Barclay WS, Johnson MR. Pregnancy-related im-
munol.1200544. mune suppression leads to altered influenza vaccine recall responses. Clin Immunol
[42] Tsuda S, Zhang X, Hamana H, Shima T, Ushijima A, Tsuda K, et al. Clonally ex- 2019;208:108254. doi:10.1016/j.clim.2019.108254.
panded decidual effector regulatory T cells increase in late gestation of normal [68] Garcia-Flores V, Romero R, Xu Y, Theis KR, Arenas-Hernandez M, Miller D, et al.
pregnancy, but not in preeclampsia, in humans. Front Immunol 2018;9:1934. Maternal-fetal immune responses in pregnant women infected with SARS-CoV-2.
doi:10.3389/fimmu.2018.01934. Nat Commun 2022;13(1):320. doi:10.1038/s41467-021-27745-z.
[43] Bai K, Lee CL, Liu X, Li J, Cao D, Zhang L, et al. Human placental exosomes induce [69] Fenizia C, Biasin M, Cetin I, Vergani P, Mileto D, Spinillo A, et al. Analysis of SARS-
maternal systemic immune tolerance by reprogramming circulating monocytes. J CoV-2 vertical transmission during pregnancy. Nat Commun 2020;11(1):5128.
Nanobiotechnol 2022;20(1):86. doi:10.1186/s12951-022-01283-2. doi:10.1038/s41467-020-18933-4.
[44] Holland OJ, Linscheid C, Hodes HC, Nauser TL, Gilliam M, Stone P, et al. Mi- [70] Joerger-Messerli MS, Hoesli IM, Rusterholz C, Lapaire O. Stimulation of mono-
nor histocompatibility antigens are expressed in syncytiotrophoblast and tro- cytes by placental microparticles involves toll-like receptors and nuclear fac-
phoblast debris: implications for maternal alloreactivity to the fetus. Am J Pathol tor kappa-light-chain-enhancer of activated B cells. Front Immunol 2014;5:173.
2012;180(1):256–66. doi:10.1016/j.ajpath.2011.09.021. doi:10.3389/fimmu.2014.00173.
[45] Mincheva-Nilsson L, Nagaeva O, Chen T, Stendahl U, Antsiferova J, Mogren I, [71] Goulopoulou S, Matsumoto T, Bomfim GF, Webb RC. Toll-like receptor 9
et al. Placenta-derived soluble MHC class I chain-related molecules down-regulate activation: a novel mechanism linking placenta-derived mitochondrial DNA
NKG2D receptor on peripheral blood mononuclear cells during human preg- and vascular dysfunction in pre-eclampsia. Clin Sci 2012;123(7):429–35.
nancy: a possible novel immune escape mechanism for fetal survival. J Immunol doi:10.1042/CS20120130.
2006;176(6):3585–92. doi:10.4049/jimmunol.176.6.3585. [72] Xiao H, Siddiqui J, Remick DG. Mechanisms of mortality in early and late sepsis.
[46] Stenqvist AC, Nagaeva O, Baranov V, Mincheva-Nilsson L. Exosomes secreted by Infect Immun 2006;74(9):5227–35. doi:10.1128/IAI.01220-05.
human placenta carry functional Fas ligand and TRAIL molecules and convey apop- [73] Okeke EB, Okwor I, Mou Z, Jia P, Uzonna JE. CD4+CD25+ regulatory T cells
tosis in activated immune cells, suggesting exosome-mediated immune privilege of attenuate lipopolysaccharide-induced systemic inflammatory responses and pro-
the fetus. J Immunol 2013;191(11):5515–23. doi:10.4049/jimmunol.1301885. motes survival in murine Escherichia coli infection. Shock 2013;40(1):65–73.
[47] McCartney SA, Kolarova T, Kanaan SB, Chae A, Laughney CI, Nelson JL, et al. doi:10.1097/SHK.0b013e318296e65b.
Increased fetal microchimerism in immune and stem cell subsets in preeclampsia. [74] Murphy TJ, Ni Choileain N, Zang Y, Mannick JA, Lederer JA. CD4+CD25+
Am J Reprod Immunol 2023;89(3):e13666. doi:10.1111/aji.13666. regulatory T cells control innate immune reactivity after injury. J Immunol
[48] Szekeres-Bartho J, Autran B, Debre P, Andreu G, Denver L, Chaouat G. Im- 2005;174(5):2957–63. doi:10.4049/jimmunol.174.5.2957.
munoregulatory effects of a suppressor factor from healthy pregnant women’s [75] Takeda J, Fang X, Olson DM. Pregnant human peripheral leukocyte migra-
lymphocytes after progesterone induction. Cell Immunol 1989;122(2):281–94. tion during several late pregnancy clinical conditions: a cross-sectional observa-
doi:10.1016/0008-8749(89)90077-4. tional study. BMC Pregnancy Childbirth 2017;17(1):16. doi:10.1186/s12884-016-
[49] Szekeres-Bartho J, Hadnagy J, Pacsa AS. The suppressive effect of progesterone 1204-5.
on lymphocyte cytotoxicity: unique progesterone sensitivity of pregnancy lympho- [76] Morita MH, Vázquez ML, Yamano LM, Kawano Jda S, Mosca T, Forte WC.
cytes. J Reprod Immunol 1985;7(2):121–8. doi:10.1016/0165-0378(85)90066-x. Functional activity of neutrophilic polymorphonuclear leukocytes in the

13
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

first five days postpartum. Rev Bras Ginecol Obstet 2015;37(11):512–15. Group B Streptococcus during pregnancy. Mol Syst Biol 2023;19(3):e11021.
doi:10.1590/SO100-720320150005434. doi:10.15252/msb.202211021.
[77] Sanghavi M, Rutherford JD. Cardiovascular physiology of pregnancy. Circulation [105] Orsaria M, Liviero S, Rossetti E, Pittini C, Driul L, Londero AP, et al. Placental acute
2014;130(12):1003–8. doi:10.1161/circulationaha.114.009029. inflammation infiltrates and pregnancy outcomes: a retrospective cohort study. Sci
[78] Taranikanti M. Physiological changes in cardiovascular system during normal preg- Rep 2021;11(1):24165. doi:10.1038/s41598-021-03655-4.
nancy: a review. Indian J Cardiovasc Dis Women-WINCARS 2018;03(02/03):62–7. [106] Delzell JE JR, Lefevre ML. Urinary tract infections during pregnancy. Am Fam
doi:10.1055/s-0038-1676666. Physician 2000;61(3):713–21.
[79] Nama V, Antonios TF, Onwude J, Manyonda IT. Mid-trimester blood pressure [107] Khalesi N, Khosravi N, Jalali A, Amini L. Evaluation of maternal urinary tract infec-
drop in normal pregnancy: myth or reality. J Hypertens 2011;29(4):763–8. tion as a potential risk factor for neonatal urinary tract infection. J Family Reprod
doi:10.1097/HJH.0b013e328342cb02. Health 2014;8(2):59–62.
[80] Kristiansson P, Wang JX. Reproductive hormones and blood pressure during preg- [108] Bilgin H, Yalinbas EE, Elifoglu I, Atlanoglu S. Maternal urinary tract infection:
nancy. Hum Reprod 2001;16(1):13–17. doi:10.1093/humrep/16.1.13. is it associated with neonatal urinary tract infection? J Family Reprod Health
[81] Birukov A, Andersen LB, Herse F, Rakova N, Kitlen G, Kyhl HB, et al. Aldos- 2021;15(1):8–12. doi:10.18502/jfrh.v15i1.6067.
terone, salt, and potassium intakes as predictors of pregnancy outcome, includ- [109] Bayih WA, Ayalew MY, Chanie ES, Abate BB, Alemayehu SA, Belay DM,
ing preeclampsia. Hypertension 2019;74(2):391–8. doi:10.1161/HYPERTENSION- et al. The burden of neonatal sepsis and its association with antenatal uri-
AHA.119.12924. nary tract infection and intra-partum fever among admitted neonates in
[82] Szmuilowicz ED, Adler GK, Williams JS, Green DE, Yao TM, Hopkins PN, et al. Ethiopia: a systematic review and meta-analysis. Heliyon 2021;7(2):e06121.
Relationship between aldosterone and progesterone in the human menstrual cycle. doi:10.1016/j.heliyon.2021.e06121.
J Clin Endocrinol Metab 2006;91(10):3981–7. doi:10.1210/jc.2006-1154. [110] Chen YH, Keller J, Wang IT, Lin CC, Lin HC. Pneumonia and pregnancy outcomes:
[83] Faúndes A, Brícola-Filho M, Pinto e Silva JL. Dilatation of the urinary tract during a nationwide population-based study. Am J Obstet Gynecol 2012;207(4):288 e1-7.
pregnancy: proposal of a curve of maximal caliceal diameter by gestational age. doi:10.1016/j.ajog.2012.08.023.
Am J Obstet Gynecol 1998;178(5):1082–6. doi:10.1016/s0002-9378(98)70552-6. [111] Allotey J, Stallings E, Bonet M, Yap M, Chatterjee S, Kew T, et al. Clinical
[84] Antonucci E, Fiaccadori E, Donadello K, Taccone FS, Franchi F, Scolletta S. Myocar- manifestations, risk factors, and maternal and perinatal outcomes of coronavirus
dial depression in sepsis: from pathogenesis to clinical manifestations and treat- disease 2019 in pregnancy: living systematic review and meta-analysis. BMJ
ment. J Crit Care 2014;29(4):500–11. doi:10.1016/j.jcrc.2014.03.028. 2020;370:m3320. doi:10.1136/bmj.m3320.
[85] Court O, Kumar A, Parrillo JE, Kumar A. Clinical review: myocardial depression in [112] Docherty AB, Harrison EM, Green CA, Hardwick HE, Pius R, Norman L, et al.
sepsis and septic shock. Crit Care 2002;6(6):500–8. doi:10.1186/cc1822. Features of 20 133 UK patients in hospital with covid-19 using the ISARIC WHO
[86] Razazi K, Boissier F, Surenaud M, Bedet A, Seemann A, Carteaux G, et al. A multi- Clinical Characterisation Protocol: prospective observational cohort study. BMJ
plex analysis of sepsis mediators during human septic shock: a preliminary study 2020;369:m1985. doi:10.1136/bmj.m1985.
on myocardial depression and organ failures. Ann Intensive Care 2019;9(1):64. [113] Mullins E, Hudak ML, Banerjee J, Getzlaff T, Townson J, Barnette K, et al.
doi:10.1186/s13613-019-0538-3. Pregnancy and neonatal outcomes of COVID-19: coreporting of common out-
[87] Klouche K, Pommet S, Amigues L, Bargnoux AS, Dupuy AM, Machado S, comes from PAN-COVID and AAP-SONPM registries. Ultrasound Obstet Gynecol
et al. Plasma brain natriuretic peptide and troponin levels in severe sep- 2021;57(4):573–81. doi:10.1002/uog.23619.
sis and septic shock: relationships with systolic myocardial dysfunction and [114] Engjom HM, Ramakrishnan R, Vousden N, Bunch K, Morris E, Simpson N, et al.
intensive care unit mortality. J Intensive Care Med 2014;29(4):229–37. Severity of maternal SARS-CoV-2 infection and perinatal outcomes of women ad-
doi:10.1177/0885066612471621. mitted to hospital during the omicron variant dominant period using UK Ob-
[88] Sato R, Nasu M. A review of sepsis-induced cardiomyopathy. J Intensive Care stetric Surveillance System data: prospective, national cohort study. BMJ Med
2015;3:48. doi:10.1186/s40560-015-0112-5. 2022;1(1):e000190. doi:10.1136/bmjmed-2022-000190.
[89] Rivers EP, McCord J, Otero R, Jacobsen G, Loomba M. Clinical utility of B-type [115] Dileep A, ZainAlAbdin S, AbuRuz S. Investigating the association between sever-
natriuretic peptide in early severe sepsis and septic shock. J Intensive Care Med ity of COVID-19 infection during pregnancy and neonatal outcomes. Sci Rep
2007;22(6):363–73. doi:10.1177/0885066607307523. 2022;12(1):3024. doi:10.1038/s41598-022-07093-8.
[90] Liu DB, Armstrong WR 3rd, Maizels M. Hydronephrosis: prenatal and [116] Gale C, Quigley MA, Placzek A, Knight M, Ladhani S, Draper ES, et al. Charac-
postnatal evaluation and management. Clin Perinatol 2014;41(3):661–78. teristics and outcomes of neonatal SARS-CoV-2 infection in the UK: a prospec-
doi:10.1016/j.clp.2014.05.013. tive national cohort study using active surveillance. Lancet Child Adolesc Health
[91] Gomez H, Ince C, De Backer D, Pickkers P, Payen D, Hotchkiss J, et al. A unified the- 2021;5(2):113–21. doi:10.1016/S2352-4642(20)30342-4.
ory of sepsis-induced acute kidney injury: inflammation, microcirculatory dysfunc- [117] Tan L, Lacko LA, Zhou T, Tomoiaga D, Hurtado R, Zhang T, et al. Pre- and peri-
tion, bioenergetics, and the tubular cell adaptation to injury. Shock 2014;41(1):3– implantation Zika virus infection impairs fetal development by targeting trophec-
11. doi:10.1097/shk.0000000000000052. toderm cells. Nat Commun 2019;10(1):4155. doi:10.1038/s41467-019-12063-2.
[92] Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol [118] Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link between
2014;5:461. doi:10.3389/fimmu.2014.00461. ACE2 deficiency and SARS-CoV-2 infection. Eur J Intern Med 2020;76:14–20.
[93] Peerapornratana S, Manrique-Caballero CL, Gómez H, Kellum JA. Acute kidney doi:10.1016/j.ejim.2020.04.037.
injury from sepsis: current concepts, epidemiology, pathophysiology, prevention [119] Gengler C, Dubruc E, Favre G, Greub G, de Leval L, Baud D. SARS-CoV-2 ACE-
and treatment. Kidney Int 2019;96(5):1083–99. doi:10.1016/j.kint.2019.05.026. receptor detection in the placenta throughout pregnancy. Clin Microbiol Infect
[94] LoMauro A, Aliverti A. Respiratory physiology of pregnancy: physiology master- 2021;27(3):489–90. doi:10.1016/j.cmi.2020.09.049.
class. Breathe 2015;11(4):297–301. doi:10.1183/20734735.008615. [120] Ryan L, Plötz FB, van den Hoogen A, Latour JM, Degtyareva M, Keuning M,
[95] Lee SY, Chien DK, Huang CH, Shih SC, Lee WC, Chang WH. Dyspnea in pregnancy. et al. Neonates and COVID-19: state of the art: neonatal sepsis series. Pediatr Res
Taiwan J Obstet Gynecol 2017;56(4):432–6. doi:10.1016/j.tjog.2017.04.035. 2022;91(2):432–9. doi:10.1038/s41390-021-01875-y.
[96] Jensen D, Wolfe LA, Slatkovska L, Webb KA, Davies GA, DE O’Donnell. Ef- [121] Argueta LB, Lacko LA, Bram Y, Tada T, Carrau L, Rendeiro AF, et al. Inflammatory
fects of human pregnancy on the ventilatory chemoreflex response to car- responses in the placenta upon SARS-CoV-2 infection late in pregnancy. iScience
bon dioxide. Am J Physiol Regul Integr Comp Physiol 2005;288(5):R1369–75. 2022;25(5):104223. doi:10.1016/j.isci.2022.104223.
doi:10.1152/ajpregu.00862.2004. [122] Lu-Culligan A, Chavan AR, Vijayakumar P, Irshaid L, Courchaine EM, Milano KM,
[97] Cérbulo-Vázquez A, Figueroa-Damián R, Arriaga-Pizano LA, Hernández-Andrade E, et al. Maternal respiratory SARS-CoV-2 infection in pregnancy is associated with a
Mancilla-Herrera I, Flores-Mejía LA, et al. Pregnant women infected with pan- robust inflammatory response at the maternal-fetal interface. Med 2021;2(5):591–
demic H1N1pdm2009 influenza virus displayed overproduction of peripheral 610 e10. doi:10.1016/j.medj.2021.04.016.
blood CD69+ lymphocytes and increased levels of serum cytokines. PLoS One [123] Faure-Bardon V, Isnard P, Roux N, Leruez-Ville M, Molina T, Bessieres B,
2014;9(9):e107900. doi:10.1371/journal.pone.0107900. et al. Protein expression of angiotensin-converting enzyme 2, a SARS-CoV-2-
[98] Bonet M, Souza JP, Abalos E, Fawole B, Knight M, Kouanda S, et al. The global specific receptor, in fetal and placental tissues throughout gestation: new in-
maternal sepsis study and awareness campaign (GLOSS): study protocol. Reprod sight for perinatal counseling. Ultrasound Obstet Gynecol 2021;57(2):242–7.
Health 2018;15(1):16. doi:10.1186/s12978-017-0437-8. doi:10.1002/uog.22178.
[99] Frascoli M, Coniglio L, Witt R, Jeanty C, Fleck-Derderian S, Myers DE, et al. Allore- [124] National Institute for Health and Care Excellence. Neonatal infection (early
active fetal T cells promote uterine contractility in preterm labor via IFN-𝛾 and TNF- onset): antibiotics for prevention and treatment [CG149]. Available from:
𝛼. Sci Transl Med 2018;10(438):eaan2263. doi:10.1126/scitranslmed.aan2263. https://www.nice.org.uk/guidance/cg149. [Accessed 22nd August 2012]
[100] Simonsen KA, Anderson-Berry AL, Delair SF, Davies HD. Early-onset neonatal sep- [125] Goel N, Shrestha S, Smith R, Mehta A, Ketty M, Muxworthy H, et al. Screening
sis. Clin Microbiol Rev 2014;27(1):21–47. doi:10.1128/CMR.00031-13. for early onset neonatal sepsis: NICE guidance-based practice versus projected
[101] Sgro M, Campbell DM, Mellor KL, Hollamby K, Bodani J, Shah PS. Early-onset application of the Kaiser Permanente sepsis risk calculator in the UK popula-
neonatal sepsis: organism patterns between 2009 and 2014. Paediatr Child Health tion. Arch Dis Child Fetal Neonatal Ed 2020;105(2):118–22. doi:10.1136/archdis-
2019;25(7):425–31. doi:10.1093/pch/pxz073. child-2018-316777.
[102] Bellizzi S, Bassat Q, Ali MM, Sobel HL, Temmerman M. Effect of puerperal [126] Watson G, Caldwell C, Kennea N. Neonatal early onset sepsis: a reflection on the
infections on early neonatal mortality: a secondary analysis of six demo- NICE guidance. Infant 2016;12(4):133–5.
graphic and health surveys. PLoS One 2017;12(1):e0170856. doi:10.1371/jour- [127] Branstetter JW, Barker L, Yarbrough A, Ross S, Stultz JS. Challenges of antibiotic
nal.pone.0170856. stewardship in the pediatric and neonatal intensive care units. J Pediatr Pharmacol
[103] Mukhopadhyay S, Puopolo KM. Risk assessment in neonatal early onset sepsis. Ther 2021;26(7):659–68. doi:10.5863/1551-6776-26.7.659.
Semin Perinatol 2012;36(6):408–15. doi:10.1053/j.semperi.2012.06.002. [128] Neonatal Early-Onset Sepsis Calculator [Internet]. [updated 2023]. Available
[104] Kuperwaser F, Avital G, Vaz MJ, Noble KN, Dammann AN, Randis TM, from: https://neonatalsepsiscalculator.kaiserpermanente.org/InfectionProbability
et al. Host inflammatory dynamics reveal placental immune modulation by Calculator.aspx.

14
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

[129] Pettinger KJ, Mayers K, McKechnie L, Phillips B. Sensitivity of the Kaiser Perma- [158] Charani E, Cunnington AJ, Yousif A, Seed Ahmed M, Ahmed A, Babiker S, et al.
nente early-onset sepsis calculator: a systematic review and meta-analysis. EClini- In transition: current health challenges and priorities in Sudan. BMJ Glob Health
calMedicine 2020;19:100227. doi:10.1016/j.eclinm.2019.11.020. 2019;4(4):e001723. doi:10.1136/bmjgh-2019-001723.
[130] Klingenberg C, Kornelisse RF, Buonocore G, Maier RF, Stocker M. Culture-negative [159] Courtenay M, Castro-Sanchez E, Fitzpatrick M, Gallagher R, Lim R, Morris G. Tack-
early-onset neonatal sepsis - at the crossroad between efficient sepsis care and an- ling antimicrobial resistance 2019-2024 - The UK’s five-year national action plan.
timicrobial stewardship. Front Pediatr 2018;6:285. doi:10.3389/fped.2018.00285. J Hosp Infect 2019;101(4):426–7. doi:10.1016/j.jhin.2019.02.019.
[131] Risnes KR, Belanger K, Murk W, Bracken MB. Antibiotic exposure by 6 months and [160] Wilkie GL, Prabhu M, Ona S, Easter SR, Tuomala RE, Riley LE, et al. Mi-
asthma and allergy at 6 years: findings in a cohort of 1,401 US children. Am J crobiology and antibiotic resistance in peripartum bacteremia. Obstet Gynecol
Epidemiol 2011;173(3):310–18. doi:10.1093/aje/kwq400. 2019;133(2):269–75. doi:10.1097/AOG.0000000000003055.
[132] Murk W, Risnes KR, Bracken MB. Prenatal or early-life exposure to antibiotics and [161] Boyd A, Lewis A, Dallas S, Xenakis E, Ramsey PS. Comparison of obstetric to institu-
risk of childhood asthma: a systematic review. Pediatrics 2011;127(6):1125–38. tional antibiograms as an approach to advance antimicrobial stewardship in mater-
doi:10.1542/peds.2010-2092. nal care. Am J Obstet Gynecol 2019;221(6):666. doi:10.1016/j.ajog.2019.10.071.
[133] World Health Organization. Antimicrobial resistance: global report on surveillance. [162] Allegranzi B., Bagheri Nejad S., Garcia G.C., Kilpatrick C., Kelley E., Mathai E. Re-
Available from: https://www.who.int/publications/i/item/9789241564748. [Ac- port on the burden of endemic health care-associated infection worldwide. Clean
cessed 1st April 2014]. care is safer care. Available from: https://www.who.int/publications/i/item/
[134] Cotten CM. Adverse consequences of neonatal antibiotic exposure. Curr Opin Pe- report-on-the-burden-of-endemic-health-care-associated-infection-worldwide.
diatr 2016;28(2):141–9. doi:10.1097/MOP.0000000000000338. [Accessed 12nd January 2011].
[135] Li G, Bielicki JA, Ahmed A, Islam MS, Berezin EN, Gallacci CB, et al. Towards [163] National Statistics. Clostridioides difficile (C. difficile) infection: an-
understanding global patterns of antimicrobial use and resistance in neonatal nual data; 2019. Available from: https://www.gov.uk/government/
sepsis: insights from the NeoAMR network. Arch Dis Child 2020;105(1):26–31. statistics/clostridium-difficile-infection-annual-data. [Accessed 7th June 2020].
doi:10.1136/archdischild-2019-316816. [164] National Statistics. MRSA bacteraemia: annual data; 2019. Available from:
[136] Rhodes A, Evans LE, Alhazzani W, Levy MM, Antonelli M, Ferrer R, https://www.gov.uk/government/statistics/mrsa-bacteraemia-annual-data. [Ac-
et al. Surviving sepsis campaign: international guidelines for management cessed 7th June 2020].
of sepsis and septic shock: 2016. Intensive Care Med 2017;43(3):304–77. [165] Hsia Y, Lee BR, Versporten A, Yang Y, Bielicki J, Jackson C, et al. Use of the WHO
doi:10.1007/s00134-017-4683-6. access, watch, and reserve classification to define patterns of hospital antibiotic
[137] Daniels R, Nutbeam T, McNamara G, Galvin C. The sepsis six and the severe sep- use (AWaRe): an analysis of paediatric survey data from 56 countries. Lancet Glob
sis resuscitation bundle: a prospective observational cohort study. Emerg Med J Health 2019;7(7):e861–ee71. doi:10.1016/s2214-109x(19)30071-3.
2011;28(6):507–12. doi:10.1136/emj.2010.095067. [166] Islam MS, Charani E, Holmes AH. The AWaRe point prevalence study index:
[138] Nutbeam T., Daniels R. On behalf of the UK sepsis trust. Available from: simplifying surveillance of antibiotic use in paediatrics. Lancet Glob Health
https://sepsistrust.org/professional-resources/clinical/. [Accessed 7th June 2020]. 2019;7(7):e811–12. doi:10.1016/S2214-109X(19)30162-7.
[139] Burston J, Adhikari S, Hayen A, Doolan H, Kelly ML, Fu K, et al. A role for antimi- [167] Antimicrobial stewardship programmes in health-care facilities in low- and
crobial stewardship in clinical sepsis pathways: a prospective interventional study. middle-income countries: a WHO practical toolkit. JAC Antimicrob Resist
Infect Control Hosp Epidemiol 2017;38(9):1032–8. doi:10.1017/ice.2017.139. 2019;1(3):dlz072. doi:10.1093/jacamr/dlz072.
[140] Prevention of Early-onset Neonatal Group B Streptococcal Disease. BJOG [168] Health and Care Excellence. Antimicrobial stewardship: systems and processes
2017;124(12):e280–305. doi: 10.1111/1471-0528.14821. for effective antimicrobial medicine use. NICE guideline [NG15]. Available from:
[141] World Health Organization. WHO recommendation on prophylactic antibiotics https://www.nice.org.uk/guidance/ng15. [Accessed 18th August 2015].
for women undergoing caesarean section. Available from: https://www.who.int/ [169] Hartman E, van de Pol AC, Heltveit-Olsen SR, Lindbæk M, Høye S, Lithén SS, et al.
publications/i/item/9789240028012. [Accessed 18th Jun 2021]. Effect of a multifaceted antibiotic stewardship intervention to improve antibiotic
[142] ACOG Practice Bulletin No. 199Use of prophylactic antibiotics in labor prescribing for suspected urinary tract infections in frail older adults (ImpresU):
and delivery. Obstet Gynecol 2018;132(3):e103–ee19. doi:10.1097/aog. pragmatic cluster randomised controlled trial in four European countries. BMJ
0000000000002833. 2023;380:e072319. doi:10.1136/bmj-2022-072319.
[143] WHO recommendation on routine antibiotic prophylaxis for women undergoing [170] Al-Omari A, Al Mutair A, Alhumaid S, Salih S, Alanazi A, Albarsan H, et al. The im-
operative vaginal birth. Geneva: World Health Organization; 2021. pact of antimicrobial stewardship program implementation at four tertiary private
[144] Murphy DJ, Strachan BK, Bahl R. The Royal College of O, Gynaecologists. Assisted hospitals: results of a five-years pre-post analysis. Antimicrob Resist Infect Control
vaginal birth. BJOG 2020;127(9):e70–e112. doi:10.1111/1471-0528.16092. 2020;9(1):95. doi:10.1186/s13756-020-00751-4.
[145] National Maternity Network: Management of intrapartum maternal pyrexia in hos- [171] Sepsis: recognition, diagnosis and early management: © NICE (2017) sepsis:
pital guideline. Scotland: NHS Scotland. Scotish Perinatal Network. Available from: recognition, diagnosis and early management. BJU Int 2018;121(4):497–514.
https://nhslguidelines.scot.nhs.uk/media/2280/management-of-intrapartum- doi:10.1111/bju.14179.
maternal-pyrexia-in-hospital-v10.pdf. [Accessed 5th September 2022]. [172] Knight M, Kenyon S, Brocklehurst P, Neilson J, Shakespeare J, editors. KJ. Saving
[146] Royal College of Obstetricians & Gynaecologists, Gynaecologists RCoOa. lives, improving mothers’ care - lessons learned to inform future maternity care
Bacterial sepsis in pregnancy. Greentop guideline No.64a. In: RCOG, ed. from the UK and Ireland confidential enquiries into maternal deaths and morbid-
Greentop Guidelines. 64a. 2012. Available from: https://www.rcog.org.uk/ ity 2009–12, Oxford: University of Oxford; 2014. National Perinatal Epidemiology
guidance/browse-all-guidance/green-top-guidelines/sepsis-in-pregnancy-bacterial Unit.
-green-top-guideline-no-64a/. [Accessed 25th April 2012]. [173] Umar A, Ameh CA, Muriithi F, Mathai M. Early warning systems in obstetrics: a
[147] Casaubon J. Mastitis and breast abscess. BMJ Best Pract 2023. March systematic literature review. PLoS One 2019;14(5) e0217864-e. doi:10.1371/jour-
https://bestpractice.bmj.com/topics/en-gb/1084 [Accessed 1st July 2023]. nal.pone.0217864.
[148] Mackeen AD, Packard RE, Ota E, Speer L. Antibiotic regimens for post- [174] Burke J, Wood S, Hermon A, Szakmany T. Improving outcome of sepsis on
partum endometritis. Cochrane Database Syst Rev 2015;2015(2):CD001067. the ward: introducing the ’Sepsis Six’ bundle. Nurs Crit Care 2019;24(1):33–9.
doi:10.1002/14651858.CD001067.pub3. doi:10.1111/nicc.12358.
[149] National Institute for Health and Care Excellence. Pyelonephritis (acute): [175] Knight M, Bunch K, Tuffnell D, Patel R, Shakespeare J, Kotnis R, et al. Saving lives,
antimicrobial prescribing. NICE guideline [NG111]. Available from: improving mothers’ care - lessons learned to inform maternity care from the UK
https://www.nice.org.uk/guidance/ng111. [Accessed 31st October 2018]. and Ireland confidential enquiries into maternal deaths and morbidity 2017-19.
[150] Long B.J. Toxic shock syndrome. BMJ best practice. May 2023. Oxford: Oxford: National Perinatal Epidemiology Unit, University of Oxford; 2021.
https://bestpractice.bmj.com/topics/en-gb/329.[Accessed 1st July 2023] [176] Benitz WE, Achten NB. Finding a role for the neonatal early-onset sepsis risk cal-
[151] National Institute for Health and Care Excellence. Neonatal infection: antibi- culator. EClinicalMedicine 2020;19:100255. doi:10.1016/j.eclinm.2019.100255.
otics for prevention and treatment. NICE guideline [NG195]. Available from: [177] Mothers and Babies: reducing risk through audits and confidential enquiries across
https://www.nice.org.uk/guidance/ng195. [Accessed 20th April 2021]. the UK. Saving lives, improving mothers’ care: lessons learned to inform mater-
[152] Murray CJL, Ikuta KS, Sharara F, Swetschinski L, Robles Aguilar G, Gray A, et al. nity care from the UK and Ireland confidential enquiries into maternal deaths and
Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. morbidity 2015–17. 2019.
Lancet 2022;399(10325):629–55. doi:10.1016/S0140-6736(21)02724-0. [178] Greer O, Shah NM, Johnson MR. Maternal sepsis update: current management and
[153] European Centre for Disease Prevention and Control. Surveillance of antimicro- controversies. Obstet Gynaecol 2020;22(1):45–55. doi:10.1111/tog.12623.
bial resistance in Europe 2018. Antimicrobial resistance surveillance in Europe. [179] Bowyer L, Robinson HL, Barrett H, Crozier TM, Giles M, Idel I, et al. SOMANZ
Available from: https://www.ecdc.europa.eu/en/publications-data/surveillance- guidelines for the investigation and management sepsis in pregnancy. Aust N Z J
antimicrobial-resistance-europe-2018. [Accessed 18th November 2019]. Obstet Gynaecol 2017;57(5):540–51. doi:10.1111/ajo.12646.
[154] Ibrahim ME, Bilal NE, Hamid ME. Increased multi-drug resistant Escherichia coli [180] Haas DM, Morgan S, Contreras K, Enders S. Vaginal preparation with antiseptic
from hospitals in Khartoum state, Sudan. Afr Health Sci 2012;12(3):368–75. solution before cesarean section for preventing postoperative infections. Cochrane
doi:10.4314/ahs.v12i3.19. Database Syst Rev 2018;7(7):CD007892. doi:10.1002/14651858.CD007892.pub6.
[155] Kheder S, Ali N, Fathelrahman A. Prevalence and antimicrobial susceptibility pat- [181] Otu A, Nsutebu EF, Hirst JE, Thompson K, Walker K, Yaya S. How to close
tern of methicillin resistance Staphylococcus in a Sudanese surgical ward. Pharma- the maternal and neonatal sepsis gap in sub-Saharan Africa. BMJ Glob Health
col Pharm 2012;3:103–8. doi:10.4236/pp.2012.31015. 2020;5(4):e002348. doi:10.1136/bmjgh-2020-002348.
[156] Kheder S, Mahmoud R, Ali H. Prescribing rationality in Khartoum State, Sudan: an [182] Steinhagen F, Schmidt SV, Schewe JC, Peukert K, Klinman DM, Bode C. Im-
update. Sudan Med J 2014;9(2):61–5. doi:10.4103/1858-5000.146575. munotherapy in sepsis - brake or accelerate. Pharmacol Ther 2020;208:107476.
[157] Oleim S, Noor S, Bushara S, Ahmed M, Elmadhoun W. The irrational use of antibi- doi:10.1016/j.pharmthera.2020.107476.
otics among doctors, pharmacists and the public in river Nile State, Sudan. Sudan [183] Peters van Ton AM, Kox M, Abdo WF, Pickkers P. Precision immunotherapy for
J Med Sci 2019;14:1–13. doi:10.18502/sjms.v14i4.5909. sepsis. Front Immunol 2018;9:1926. doi:10.3389/fimmu.2018.01926.

15
JID: JOINTM
ARTICLE IN PRESS [m5GeSdc;September 14, 2023;5:26]

N.M. Shah, E. Charani, D. Ming et al. Journal of Intensive Medicine xxx (xxxx) xxx

[184] Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, et al. Dexametha- with intact membranes. Am J Obstet Gynecol 2014;210(2) 125.e1-125.e15.
sone in hospitalized patients with Covid-19. N Engl J Med 2021;384(8):693–704. doi:10.1016/j.ajog.2013.11.032.
doi:10.1056/NEJMoa2021436. [203] Urushiyama D, Suda W, Ohnishi E, Araki R, Kiyoshima C, Kurakazu M, et al. Micro-
[185] Abani O, Abbas A, Abbas F, Abbas M, Abbasi S, Abbass H, et al. biome profile of the amniotic fluid as a predictive biomarker of perinatal outcome.
Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): a ran- Sci Rep 2017;7(1):12171. doi:10.1038/s41598-017-11699-8.
domised, controlled, open-label, platform trial. Lancet 2021;397(10285):1637–45. [204] Stinson L, Hallingström M, Barman M, Viklund F, Keelan J, Kacerovsky M,
doi:10.1016/S0140-6736(21)00676-0. et al. Comparison of bacterial DNA profiles in mid-trimester amniotic fluid
[186] Bo L, Wang F, Zhu J, Li J, Deng X. Granulocyte-colony stimulating factor (G- samples from preterm and term deliveries. Front Microbiol 2020;11:415.
CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF) for sepsis: doi:10.3389/fmicb.2020.00415.
a meta-analysis. Crit Care 2011;15(1):R58. doi:10.1186/cc10031. [205] Kenyon SL, Taylor DJ, Tarnow-Mordi W. Broad-spectrum antibiotics for
[187] Cui J, Wei X, Lv H, Li Y, Li P, Chen Z, et al. The clinical efficacy of intra- spontaneous preterm labour: the ORACLE II randomised trial. Lancet
venous IgM-enriched immunoglobulin (pentaglobin) in sepsis or septic shock: a 2001;357(9261):989–94. doi:10.1016/S0140-6736(00)04234-3.
meta-analysis with trial sequential analysis. Ann Intensive Care 2019;9(1):27. [206] Kenyon SL, Taylor DJ, Tarnow-Mordi W. Broad-spectrum antibiotics for
doi:10.1186/s13613-019-0501-3. preterm, prelabour rupture of fetal membranes: the ORACLE I randomised
[188] Delsing CE, Gresnigt MS, Leentjens J, Preijers F, Frager FA, Kox M, et al. Interferon- trial. ORACLE Collaborative Group. Lancet 2001;357(9261):979–88.
gamma as adjunctive immunotherapy for invasive fungal infections: a case series. doi:10.1016/s0140-6736(00)04233-1.
BMC Infect Dis 2014;14(1):166. doi:10.1186/1471-2334-14-166. [207] Kenyon S, Taylor DJ, Tarnow-Mordi WO. ORACLE–antibiotics for preterm
[189] Venet F, Foray AP, Villars-Méchin A, Malcus C, Poitevin-Later F, Lepape A, et al. IL- prelabour rupture of the membranes: short-term and long-term outcomes. Acta
7 restores lymphocyte functions in septic patients. J Immunol 2012;189(10):5073– Paediatr Suppl 2002;91(437):12–15. doi:10.1111/j.1651-2227.2002.tb00153.x.
81. doi:10.4049/jimmunol.1202062. [208] Kenyon S, Pike K, Jones DR, Brocklehurst P, Marlow N, Salt A, et al. Child-
[190] Romee R, Cooley S, Berrien-Elliott MM, Westervelt P, Verneris MR, Wagner JE, hood outcomes after prescription of antibiotics to pregnant women with spon-
et al. First-in-human phase 1 clinical study of the IL-15 superagonist complex taneous preterm labour: 7-year follow-up of the ORACLE II trial. Lancet
ALT-803 to treat relapse after transplantation. Blood 2018;131(23):2515–27. 2008;372(9646):1319–27. doi:10.1016/s0140-6736(08)61203-9.
doi:10.1182/blood-2017-12-823757. [209] Russell AR, Steer PJ. Antibiotics in preterm labour–the ORACLE speaks. Lancet
[191] Francois B, Jeannet R, Daix T, Walton AH, Shotwell MS, Unsinger J, et al. 2008;372(9646):1276–8. doi:10.1016/S0140-6736(08)61248-9.
Interleukin-7 restores lymphocytes in septic shock: the IRIS-7 randomized clinical [210] Kenyon S, Boulvain M, Neilson JP. Antibiotics for preterm rupture
trial. JCI Insight 2018;3(5):e98960. doi:10.1172/jci.insight.98960. of membranes. Cochrane Database Syst Rev 2013(12):CD001058.
[192] Youngstein T, Hoffmann P, Gül A, Lane T, Williams R, Rowczenio DM, et al. Inter- doi:10.1002/14651858.CD001058.pub3.
national multi-centre study of pregnancy outcomes with interleukin-1 inhibitors. [211] Cappelletti M, Presicce P, Feiyang M, Senthamaraikannan P, Miller LA, Pelle-
Rheumatology 2017;56(12):2102–8. doi:10.1093/rheumatology/kex305. grini M, et al. The induction of preterm labor in rhesus macaques is deter-
[193] Shakoory B, Carcillo JA, Chatham WW, Amdur RL, Zhao H, Dinarello CA, et al. mined by the strength of immune response to intrauterine infection. PLoS Biol
Interleukin-1 receptor blockade is associated with reduced mortality in sepsis pa- 2021;19(9):e3001385. doi:10.1371/journal.pbio.3001385.
tients with features of macrophage activation syndrome: reanalysis of a prior phase [212] Yoo JY, Rho M, You YA, Kwon EJ, Kim MH, Kym S, et al. 16S rRNA gene-based
III trial. Crit Care Med 2016;44(2):275–81. doi:10.1097/ccm.0000000000001402. metagenomic analysis reveals differences in bacteria-derived extracellular vesicles
[194] Boomer JS, To K, Chang KC, Takasu O, Osborne DF, Walton AH, et al. Im- in the urine of pregnant and non-pregnant women. Exp Mol Med 2016;48(2):e208.
munosuppression in patients who die of sepsis and multiple organ failure. JAMA doi:10.1038/emm.2015.110.
2011;306(23):2594–605. doi:10.1001/jama.2011.1829. [213] Young BC, Dodge LE, Gupta M, Rhee JS, Hacker MR. Evaluation of a rapid, real-
[195] Lv S, Han M, Yi R, Kwon S, Dai C, Wang R. Anti-TNF-𝛼 therapy for patients time intrapartum group B streptococcus assay. Am J Obstet Gynecol 2011;205(4)
with sepsis: a systematic meta-analysis. Int J Clin Pract 2014;68(4):520–8. 372.e1-6. doi:10.1016/j.ajog.2011.06.087.
doi:10.1111/ijcp.12382. [214] Yang L, Haidar G, Zia H, Nettles R, Qin S, Wang X, et al. Metagenomic
[196] Richter A, Listing J, Schneider M, Klopsch T, Kapelle A, Kaufmann J, et al. Impact identification of severe pneumonia pathogens in mechanically-ventilated pa-
of treatment with biologic DMARDs on the risk of sepsis or mortality after serious tients: a feasibility and clinical validity study. Respir Res 2019;20(1):265.
infection in patients with rheumatoid arthritis. Ann Rheum Dis 2016;75(9):1667– doi:10.1186/s12931-019-1218-4.
73. doi:10.1136/annrheumdis-2015-207838. [215] Taxt AM, Avershina E, Frye SA, Naseer U, Ahmad R. Rapid identification of
[197] Madhi SA, Cutland CL, Jose L, Koen A, Govender N, Wittke F, et al. Safety and im- pathogens, antibiotic resistance genes and plasmids in blood cultures by nanopore
munogenicity of an investigational maternal trivalent group B streptococcus vac- sequencing. Sci Rep 2020;10(1):7622. doi:10.1038/s41598-020-64616-x.
cine in healthy women and their infants: a randomised phase 1b/2 trial. Lancet [216] Chaemsaithong P, Romero R, Korzeniewski SJ, Martinez-Varea A, Dong Z, Yoon BH,
Infect Dis 2016;16(8):923–34. doi:10.1016/s1473-3099(16)00152-3. et al. A point of care test for interleukin-6 in amniotic fluid in preterm prela-
[198] Zhu L, Luo F, Hu W, Han Y, Wang Y, Zheng H, et al. Bacterial communi- bor rupture of membranes: a step toward the early treatment of acute intra-
ties in the womb during healthy pregnancy. Front Microbiol 2018;9:2163. amniotic inflammation/infection. J Matern Fetal Neonatal Med 2016;29(3):360–7.
doi:10.3389/fmicb.2018.02163. doi:10.3109/14767058.2015.1006621.
[199] Wang J, Li Z, Ma X, Du L, Jia Z, Cui X, et al. Translocation of vaginal micro- [217] Park H, Park KH, Kim YM, Kook SY, Jeon SJ, Yoo H-N. Plasma inflammatory and
biota is involved in impairment and protection of uterine health. Nat Commun immune proteins as predictors of intra-amniotic infection and spontaneous preterm
2021;12(1):4191. doi:10.1038/s41467-021-24516-8. delivery in women with preterm labor: a retrospective study. BMC Pregnancy Child-
[200] Stinson LF, Boyce MC, Payne MS, Keelan JA. The not-so-sterile womb: evi- birth 2018;18(1):146. doi:10.1186/s12884-018-1780-7.
dence that the human fetus is exposed to bacteria prior to birth. Front Microbiol [218] Premature rupture membranes and tPTL: a personalised approach (PROMPT);
2019;10(1124). doi:10.3389/fmicb.2019.01124. 2021. Available from: https://clinicaltrials.gov/ct2/show/NCT04878978. [Ac-
[201] He Q, Kwok L-Y, Xi X, Zhong Z, Ma T, Xu H, et al. The meconium mi- cessed 23rd January 2022].
crobiota shares more features with the amniotic fluid microbiota than the [219] Greer O, Shah NM, Sriskandan S, Johnson MR. Sepsis: precision-based
maternal fecal and vaginal microbiota. Gut Microbes 2020;12(1):1794266. medicine for pregnancy and the puerperium. Int J Mol Sci 2019;20(21):5388.
doi:10.1080/19490976.2020.1794266. doi:10.3390/ijms20215388.
[202] Combs CA, Gravett M, Garite TJ, Hickok DE, Lapidus J, Porreco R, et al.
Amniotic fluid infection, inflammation, and colonization in preterm labor

16

You might also like